1
|
English LA, Taylor RJ, Cameron CJ, Broker EA, Dent EW. F-BAR proteins CIP4 and FBP17 function in cortical neuron radial migration and process outgrowth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620310. [PMID: 39484544 PMCID: PMC11527352 DOI: 10.1101/2024.10.25.620310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Neurite initiation from newly born neurons is a critical step in neuronal differentiation and migration. Neuronal migration in the developing cortex is accompanied by dynamic extension and retraction of neurites as neurons progress through bipolar and multipolar states. However, there is a relative lack of understanding regarding how the dynamic extension and retraction of neurites is regulated during neuronal migration. In recent work we have shown that CIP4, a member of the F-BAR family of membrane bending proteins, inhibits cortical neurite formation in culture, while family member FBP17 induces premature neurite outgrowth. These results beg the question of how CIP4 and FBP17 function in radial neuron migration and differentiation in vivo, including the timing and manner of neurite extension and retraction. Indeed, the regulation of neurite outgrowth is essential for the transitions between bipolar and multipolar states during radial migration. To examine the effects of modulating expression of CIP4 and FBP17 in vivo, we used in utero electroporation, in combination with our published Double UP technique, to compare knockdown or overexpression cells with control cells within the same mouse tissue of either sex. We show that either knockdown or overexpression of CIP4 and FBP17 results in the marked disruption of radial neuron migration by modulating neuronal morphology and neurite outgrowth, consistent with our findings in culture. Our results demonstrate that the F-BAR proteins CIP4 and FBP17 are essential for proper radial migration in the developing cortex and thus play a key role in cortical development.
Collapse
Affiliation(s)
- Lauren A English
- Neuroscience Training Program, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| | - Russell J Taylor
- Neuroscience Department, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| | - Connor J Cameron
- Neuroscience Department, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| | - Emily A Broker
- Neuroscience Department, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| | - Erik W Dent
- Neuroscience Department, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705
| |
Collapse
|
2
|
O'Donoghue L, Smolenski A. Roles of G proteins and their GTPase-activating proteins in platelets. Biosci Rep 2024; 44:BSR20231420. [PMID: 38808367 PMCID: PMC11139668 DOI: 10.1042/bsr20231420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| | - Albert Smolenski
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| |
Collapse
|
3
|
Blake TCA, Fox HM, Urbančič V, Ravishankar R, Wolowczyk A, Allgeyer ES, Mason J, Danuser G, Gallop JL. Filopodial protrusion driven by density-dependent Ena-TOCA-1 interactions. J Cell Sci 2024; 137:jcs261057. [PMID: 38323924 PMCID: PMC11006392 DOI: 10.1242/jcs.261057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
Filopodia are narrow actin-rich protrusions with important roles in neuronal development where membrane-binding adaptor proteins, such as I-BAR- and F-BAR-domain-containing proteins, have emerged as upstream regulators that link membrane interactions to actin regulators such as formins and proteins of the Ena/VASP family. Both the adaptors and their binding partners are part of diverse and redundant protein networks that can functionally compensate for each other. To explore the significance of the F-BAR domain-containing neuronal membrane adaptor TOCA-1 (also known as FNBP1L) in filopodia we performed a quantitative analysis of TOCA-1 and filopodial dynamics in Xenopus retinal ganglion cells, where Ena/VASP proteins have a native role in filopodial extension. Increasing the density of TOCA-1 enhances Ena/VASP protein binding in vitro, and an accumulation of TOCA-1, as well as its coincidence with Ena, correlates with filopodial protrusion in vivo. Two-colour single-molecule localisation microscopy of TOCA-1 and Ena supports their nanoscale association. TOCA-1 clusters promote filopodial protrusion and this depends on a functional TOCA-1 SH3 domain and activation of Cdc42, which we perturbed using the small-molecule inhibitor CASIN. We propose that TOCA-1 clusters act independently of membrane curvature to recruit and promote Ena activity for filopodial protrusion.
Collapse
Affiliation(s)
- Thomas C. A. Blake
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Helen M. Fox
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Vasja Urbančič
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Roshan Ravishankar
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam Wolowczyk
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Edward S. Allgeyer
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Julia Mason
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jennifer L. Gallop
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| |
Collapse
|
4
|
Uppakara K, Jamornwan S, Duan LX, Yue KR, Sunrat C, Dent EW, Wan SB, Saengsawang W. Novel α-Lipoic Acid/3- n-Butylphthalide Conjugate Enhances Protective Effects against Oxidative Stress and 6-OHDA Induced Neuronal Damage. ACS Chem Neurosci 2020; 11:1634-1642. [PMID: 32374999 DOI: 10.1021/acschemneuro.0c00105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative diseases are irreversible conditions that result in progressive degeneration and death of nerve cells. Although the underlying mechanisms may vary, oxidative stress is considered to be one of the major causes of neuronal loss. Importantly, there are still no comprehensive treatments to completely cure these diseases. Therefore, protecting neurons from oxidative damage may be the most effective therapeutic strategy. Here we report a neuroprotective effects of a novel hybrid compound (dlx-23), obtained by conjugating α-lipoic acid (ALA), a natural antioxidant agent, and 3-n-butylphthalide (NBP), a clinical anti-ischemic drug. Dlx-23 protected against neuronal death induced by both H2O2 induced oxidative stress in Cath.-a-differentiated (CAD) cells and 6-OHDA, a toxin model of Parkinson's disease (PD) in SH-SY5Y cells. These activities proved to be more potent than the parent compound (ALA) alone. Dlx-23 scavenged free radicals, increased glutathione levels, and prevented mitochondria damage. In addition, live imaging of primary cortical neurons demonstrated that dlx-23 protected against neuronal growth cone damage induced by H2O2. Taken together these results suggest that dlx-23 has substantial potential to be further developed into a novel neuroprotective agent against oxidative damage and toxin induced neurodegeneration.
Collapse
Affiliation(s)
- Kwanchanok Uppakara
- Toxicology Graduate Program; Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Sopana Jamornwan
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Liang-xing Duan
- Qingdao National Laboratory for Marine Science and Technology; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Kai-rui Yue
- Qingdao National Laboratory for Marine Science and Technology; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Chotchanit Sunrat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Erik W Dent
- Department of Neuroscience, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| | - Sheng-biao Wan
- Qingdao National Laboratory for Marine Science and Technology; School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Witchuda Saengsawang
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center for Neuroscience, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Excellent Center for Drug Discovery (ECDD), Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
5
|
Tonucci FM, Almada E, Borini-Etichetti C, Pariani A, Hidalgo F, Rico MJ, Girardini J, Favre C, Goldenring JR, Menacho-Marquez M, Larocca MC. Identification of a CIP4 PKA phosphorylation site involved in the regulation of cancer cell invasiveness and metastasis. Cancer Lett 2019; 461:65-77. [DOI: 10.1016/j.canlet.2019.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/08/2019] [Accepted: 07/11/2019] [Indexed: 01/08/2023]
|
6
|
Taylor KL, Taylor RJ, Richters KE, Huynh B, Carrington J, McDermott ME, Wilson RL, Dent EW. Opposing functions of F-BAR proteins in neuronal membrane protrusion, tubule formation, and neurite outgrowth. Life Sci Alliance 2019; 2:2/3/e201800288. [PMID: 31160379 PMCID: PMC6549137 DOI: 10.26508/lsa.201800288] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023] Open
Abstract
Neurite formation is a fundamental antecedent to axon and dendrite formation, but the mechanisms that underlie this important process are poorly characterized. Here, we demonstrate that two F-BAR proteins, CIP4 and FBP17, have opposing functions in early cortical neuron development. The F-BAR family of proteins play important roles in many cellular processes by regulating both membrane and actin dynamics. The CIP4 family of F-BAR proteins is widely recognized to function in endocytosis by elongating endocytosing vesicles. However, in primary cortical neurons, CIP4 concentrates at the tips of extending lamellipodia and filopodia and inhibits neurite outgrowth. Here, we report that the highly homologous CIP4 family member, FBP17, induces tubular structures in primary cortical neurons and results in precocious neurite formation. Through domain swapping and deletion experiments, we demonstrate that a novel polybasic region between the F-BAR and HR1 domains is required for membrane bending. Moreover, the presence of a poly-PxxP region in longer splice isoforms of CIP4 and FBP17 largely reverses the localization and function of these proteins. Thus, CIP4 and FBP17 function as an antagonistic pair to fine-tune membrane protrusion, endocytosis, and neurite formation during early neuronal development.
Collapse
Affiliation(s)
- Kendra L Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Russell J Taylor
- University of Wisconsin-Madison, Neuroscience Training Program, Madison, WI, USA
| | - Karl E Richters
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Brandon Huynh
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Justin Carrington
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Maeve E McDermott
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Rebecca L Wilson
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| | - Erik W Dent
- University of Wisconsin-Madison, Department of Neuroscience, Madison, WI, USA
| |
Collapse
|
7
|
The Microtubule-Associated Protein Tau Mediates the Organization of Microtubules and Their Dynamic Exploration of Actin-Rich Lamellipodia and Filopodia of Cortical Growth Cones. J Neurosci 2017; 38:291-307. [PMID: 29167405 DOI: 10.1523/jneurosci.2281-17.2017] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/10/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022] Open
Abstract
Proper organization and dynamics of the actin and microtubule (MT) cytoskeleton are essential for growth cone behaviors during axon growth and guidance. The MT-associated protein tau is known to mediate actin/MT interactions in cell-free systems but the role of tau in regulating cytoskeletal dynamics in living neurons is unknown. We used cultures of cortical neurons from postnatal day (P)0-P2 golden Syrian hamsters (Mesocricetus auratus) of either sex to study the role of tau in the organization and dynamics of the axonal growth cone cytoskeleton. Here, using super resolution microscopy of fixed growth cones, we found that tau colocalizes with MTs and actin filaments and is also located at the interface between actin filament bundles and dynamic MTs in filopodia, suggesting that tau links these two cytoskeletons. Live cell imaging in concert with shRNA tau knockdown revealed that reducing tau expression disrupts MT bundling in the growth cone central domain, misdirects trajectories of MTs in the transition region and prevents single dynamic MTs from extending into growth cone filopodia along actin filament bundles. Rescue experiments with human tau expression restored MT bundling, MT penetration into the growth cone periphery and close MT apposition to actin filaments in filopodia. Importantly, we found that tau knockdown reduced axon outgrowth and growth cone turning in Wnt5a gradients, likely due to disorganized MTs that failed to extend into the peripheral domain and enter filopodia. These results suggest an important role for tau in regulating cytoskeletal organization and dynamics during growth cone behaviors.SIGNIFICANCE STATEMENT Growth cones are the motile tips of growing axons whose guidance behaviors require interaction of the dynamic actin and microtubule cytoskeleton. Tau is a microtubule-associated protein that stabilizes microtubules in neurons and in cell-free systems regulates actin-microtubule interaction. Here, using super resolution microscopy, live-cell imaging, and tau knockdown, we show for the first time in living axonal growth cones that tau is important for microtubule bundling and microtubule exploration of the actin-rich growth cone periphery. Importantly tau knockdown reduced axon outgrowth and growth cone turning, due to disorganized microtubules that fail to enter filopodia and co-align with actin filaments. Understanding normal tau functions will be important for identifying mechanisms of tau in neurodegenerative diseases such as Alzheimer's.
Collapse
|
8
|
Patel S, Fok SYY, Stefen H, Tomanić T, Parić E, Herold R, Brettle M, Djordjevic A, Fath T. Functional characterisation of filamentous actin probe expression in neuronal cells. PLoS One 2017; 12:e0187979. [PMID: 29145435 PMCID: PMC5690639 DOI: 10.1371/journal.pone.0187979] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/30/2017] [Indexed: 11/19/2022] Open
Abstract
Genetically encoded filamentous actin probes, Lifeact, Utrophin and F-tractin, are used as tools to label the actin cytoskeleton. Recent evidence in several different cell types indicates that these probes can cause changes in filamentous actin dynamics, altering cell morphology and function. Although these probes are commonly used to visualise actin dynamics in neurons, their effects on axonal and dendritic morphology has not been systematically characterised. In this study, we quantitatively analysed the effect of Lifeact, Utrophin and F-tractin on neuronal morphogenesis in primary hippocampal neurons. Our data show that the expression of actin-tracking probes significantly impacts on axonal and dendrite growth these neurons. Lifeact-GFP expression, under the control of a pBABE promoter, caused a significant decrease in total axon length, while another Lifeact-GFP expression, under the control of a CAG promoter, decreased the length and complexity of dendritic trees. Utr261-EGFP resulted in increased dendritic branching but Utr230-EGFP only accumulated in cell soma, without labelling any neurites. Lifeact-7-mEGFP and F-tractin-EGFP in a pEGFP-C1 vector, under the control of a CMV promoter, caused only minor changes in neuronal morphology as detected by Sholl analysis. The results of this study demonstrate the effects that filamentous actin tracking probes can have on the axonal and dendritic compartments of neuronal cells and emphasise the care that must be taken when interpreting data from experiments using these probes.
Collapse
Affiliation(s)
- Shrujna Patel
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Sandra Y. Y. Fok
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Holly Stefen
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
- Neuron Culture Core Facility (NCCF), University of New South Wales, Sydney, New South Wales, Australia
| | - Tamara Tomanić
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Esmeralda Parić
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Rosanna Herold
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Merryn Brettle
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Aleksandra Djordjevic
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Thomas Fath
- Neurodegeneration and Repair Unit (NRU), School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
- Neuron Culture Core Facility (NCCF), University of New South Wales, Sydney, New South Wales, Australia
- * E-mail:
| |
Collapse
|
9
|
Meng DF, Xie P, Peng LX, Sun R, Luo DH, Chen QY, Lv X, Wang L, Chen MY, Mai HQ, Guo L, Guo X, Zheng LS, Cao L, Yang JP, Wang MY, Mei Y, Qiang YY, Zhang ZM, Yun JP, Huang BJ, Qian CN. CDC42-interacting protein 4 promotes metastasis of nasopharyngeal carcinoma by mediating invadopodia formation and activating EGFR signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:21. [PMID: 28129778 PMCID: PMC5273811 DOI: 10.1186/s13046-016-0483-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/23/2016] [Indexed: 01/08/2023]
Abstract
Background Nasopharyngeal carcinoma (NPC) is a common malignancy in Southern China and Southeast Asia. In this study, we investigated the functional and molecular mechanisms by which CDC42-interacting protein 4 (CIP4) influences NPC. Methods The expression levels of CIP4 were examined by Western blot, qRT-PCR or IHC. MTT assay was used to detect the proliferative rate of NPC cells. The invasive abilities were examined by matrigel and transwell assay. The metastatic abilities of NPC cells were revealed in BALB/c nude mice. Results We report that CIP4 is required for NPC cell motility and invasion. CIP4 promotes the activation of N-WASP that controls invadopodia formation and activates EGFR signaling, which induces downstream MMP2 (matrix metalloproteinase 2) upregulation. In addition, CIP4 could promote NPC metastasis by activating the EGFR pathway. In nude mouse models, distant metastasis was significantly inhibited in CIP4-silenced groups. High CIP4 expression is an independent adverse prognostic factor of overall survival (OS) and distant metastasis-free survival (DMFS). Conclusion We identify the critical role of CIP4 in metastasis of NPC which suggest that CIP4 may be a potential therapeutic target of NPC patients.
Collapse
Affiliation(s)
- Dong-Fang Meng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ping Xie
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Xia Peng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Rui Sun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Dong-Hua Luo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiu-Yan Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xing Lv
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lin Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ming-Yuan Chen
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Ling Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiang Guo
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Sheng Zheng
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Li Cao
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jun-Ping Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Meng-Yao Wang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Radiotherapy Department, Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, 510095, China
| | - Yan Mei
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yuan-Yuan Qiang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zi-Meng Zhang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Bi-Jun Huang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
10
|
Luo W, Lieu ZZ, Manser E, Bershadsky AD, Sheetz MP. Formin DAAM1 Organizes Actin Filaments in the Cytoplasmic Nodal Actin Network. PLoS One 2016; 11:e0163915. [PMID: 27760153 PMCID: PMC5070803 DOI: 10.1371/journal.pone.0163915] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 09/17/2016] [Indexed: 01/13/2023] Open
Abstract
A nodal cytoplasmic actin network underlies actin cytoplasm cohesion in the absence of stress fibers. We previously described such a network that forms upon Latrunculin A (LatA) treatment, in which formin DAAM1 was localized at these nodes. Knock down of DAAM1 reduced the mobility of actin nodes but the nodes remained. Here we have investigated DAAM1 containing nodes after LatA washout. DAAM1 was found to be distributed between the cytoplasm and the plasma membrane. The membrane binding likely occurs through an interaction with lipid rafts, but is not required for F-actin assembly. Interesting the forced interaction of DAAM1 with plasma membrane through a rapamycin-dependent linkage, enhanced F-actin assembly at the cell membrane (compared to the cytoplasm) after the LatA washout. However, immediately after addition of both rapamycin and LatA, the cytoplasmic actin nodes formed transiently, before DAAM1 moved to the membrane. This was consistent with the idea that DAAM1 was initially anchored to cytoplasmic actin nodes. Further, photoactivatable tracking of DAAM1 showed DAAM1 was immobilized at these actin nodes. Thus, we suggest that DAAM1 organizes actin filaments into a nodal complex, and such nodal complexes seed actin network recovery after actin depolymerization.
Collapse
Affiliation(s)
- Weiwei Luo
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Zi Zhao Lieu
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Ed Manser
- sGSK Group, Institute of Molecular and Cell Biology, Agency for Science Technology and Research, Proteos Building, 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Alexander D. Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Michael P. Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Department of Biological Sciences, Columbia University, New York, New York, 10027, United States of America
- * E-mail:
| |
Collapse
|
11
|
Forbes-Lorman RM, Harris MA, Chang WS, Dent EW, Nordheim EV, Franzen MA. Physical models have gender-specific effects on student understanding of protein structure-function relationships. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 44:326-335. [PMID: 26923186 PMCID: PMC4936926 DOI: 10.1002/bmb.20956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 12/22/2015] [Accepted: 01/12/2016] [Indexed: 05/31/2023]
Abstract
Understanding how basic structural units influence function is identified as a foundational/core concept for undergraduate biological and biochemical literacy. It is essential for students to understand this concept at all size scales, but it is often more difficult for students to understand structure-function relationships at the molecular level, which they cannot as effectively visualize. Students need to develop accurate, 3-dimensional mental models of biomolecules to understand how biomolecular structure affects cellular functions at the molecular level, yet most traditional curricular tools such as textbooks include only 2-dimensional representations. We used a controlled, backward design approach to investigate how hand-held physical molecular model use affected students' ability to logically predict structure-function relationships. Brief (one class period) physical model use increased quiz score for females, whereas there was no significant increase in score for males using physical models. Females also self-reported higher learning gains in their understanding of context-specific protein function. Gender differences in spatial visualization may explain the gender-specific benefits of physical model use observed. © 2016 The Authors Biochemistry and Molecular Biology Education published by Wiley Periodicals, Inc. on behalf of International Union of Biochemistry and Molecular Biology, 44(4):326-335, 2016.
Collapse
Affiliation(s)
| | - Michelle A Harris
- Biology Core Curriculum, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wesley S Chang
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Erik W Dent
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin
| | - Erik V Nordheim
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Margaret A Franzen
- Center for Biomolecular Modeling, Milwaukee School of Engineering, Milwaukee, Wisconsin
| |
Collapse
|
12
|
Abstract
As cells grow, move, and divide, they must reorganize and rearrange their membranes and cytoskeleton. The F-BAR protein family links cellular membranes with actin cytoskeletal rearrangements in processes including endocytosis, cytokinesis, and cell motility. Here we review emerging information on mechanisms of F-BAR domain oligomerization and membrane binding, and how these activities are coordinated with additional domains to accomplish scaffolding and signaling functions.
Collapse
Affiliation(s)
- Nathan A McDonald
- a Department of Cell and Developmental Biology , Vanderbilt University , Nashville , TN , USA
| | - Kathleen L Gould
- a Department of Cell and Developmental Biology , Vanderbilt University , Nashville , TN , USA
| |
Collapse
|
13
|
Cerqueira OLD, Truesdell P, Baldassarre T, Vilella-Arias SA, Watt K, Meens J, Chander H, Osório CAB, Soares FA, Reis EM, Craig AWB. CIP4 promotes metastasis in triple-negative breast cancer and is associated with poor patient prognosis. Oncotarget 2016; 6:9397-408. [PMID: 25823823 PMCID: PMC4496225 DOI: 10.18632/oncotarget.3351] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/10/2015] [Indexed: 01/05/2023] Open
Abstract
Signaling via epidermal growth factor receptor (EGFR) and Src kinase pathways promote triple-negative breast cancer (TNBC) cell invasion and tumor metastasis. Here, we address the role of Cdc42-interacting protein-4 (CIP4) in TNBC metastasis in vivo, and profile CIP4 expression in human breast cancer patients. In human TNBC cells, CIP4 knock-down (KD) led to less sustained activation of Erk kinase and impaired cell motility compared to control cells. This correlated with significant defects in 3D invasion of surrounding extracellular matrix by CIP4 KD TNBC cells when grown as spheroid colonies. In mammary orthotopic xenograft assays using both human TNBC cells (MDA-MB-231, HCC 1806) and rat MTLn3 cells, CIP4 silencing had no overt effect on tumor growth, but significantly reduced the incidence of lung metastases in each tumor model. In human invasive breast cancers, high CIP4 levels was significantly associated with high tumor stage, TNBC and HER2 subtypes, and risk of progression to metastatic disease. Together, these results implicate CIP4 in promoting metastasis in TNBCs.
Collapse
Affiliation(s)
- Otto L D Cerqueira
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Tomas Baldassarre
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Santiago A Vilella-Arias
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Kathleen Watt
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Jalna Meens
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Harish Chander
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| | - Cynthia A B Osório
- Department of Anatomic Pathology, A.C. Camargo Hospital, São Paulo, SP, Brazil
| | - Fernando A Soares
- Department of Anatomic Pathology, A.C. Camargo Hospital, São Paulo, SP, Brazil.,Instituto Nacional de Ciência e Tecnologia em Oncogenômica, São Paulo, SP, Brazil
| | - Eduardo M Reis
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil.,Instituto Nacional de Ciência e Tecnologia em Oncogenômica, São Paulo, SP, Brazil
| | - Andrew W B Craig
- Department of Biomedical and Molecular Sciences, Queen's University, and Division of Cancer Biology & Genetics, Queen's Cancer Research Institute, Kingston, ON, Canada
| |
Collapse
|
14
|
Nagy Z, Wynne K, von Kriegsheim A, Gambaryan S, Smolenski A. Cyclic Nucleotide-dependent Protein Kinases Target ARHGAP17 and ARHGEF6 Complexes in Platelets. J Biol Chem 2015; 290:29974-83. [PMID: 26507661 DOI: 10.1074/jbc.m115.678003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Indexed: 01/01/2023] Open
Abstract
Endothelial cells release prostacyclin (PGI2) and nitric oxide (NO) to inhibit platelet functions. PGI2 and NO effects are mediated by cyclic nucleotides, cAMP- and cGMP-dependent protein kinases (PKA, PKG), and largely unknown PKA and PKG substrate proteins. The small G-protein Rac1 plays a key role in platelets and was suggested to be a target of cyclic nucleotide signaling. We confirm that PKA and PKG activation reduces Rac1-GTP levels. Screening for potential mediators of this effect resulted in the identification of the Rac1-specific GTPase-activating protein ARHGAP17 and the guanine nucleotide exchange factor ARHGEF6 as new PKA and PKG substrates in platelets. We mapped the PKA/PKG phosphorylation sites to serine 702 on ARHGAP17 using Phos-tag gels and to serine 684 on ARHGEF6. We show that ARHGAP17 binds to the actin-regulating CIP4 protein in platelets and that Ser-702 phosphorylation interferes with this interaction. Reduced CIP4 binding results in enhanced inhibition of cell migration by ARHGAP17. Furthermore, we show that ARHGEF6 is constitutively linked to GIT1, a GAP of Arf family small G proteins, and that ARHGEF6 phosphorylation enables binding of the 14-3-3 adaptor protein to the ARHGEF6/GIT1 complex. PKA and PKG induced rearrangement of ARHGAP17- and ARHGEF6-associated protein complexes might contribute to Rac1 regulation and platelet inhibition.
Collapse
Affiliation(s)
- Zoltan Nagy
- From the UCD Conway Institute and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kieran Wynne
- Mass Spectrometry Resource, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Prospect, St. Petersburg, 194223 Russia
| | - Albert Smolenski
- From the UCD Conway Institute and the School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland,
| |
Collapse
|
15
|
Suetsugu S, Kurisu S, Takenawa T. Dynamic shaping of cellular membranes by phospholipids and membrane-deforming proteins. Physiol Rev 2014; 94:1219-48. [PMID: 25287863 DOI: 10.1152/physrev.00040.2013] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
All cellular compartments are separated from the external environment by a membrane, which consists of a lipid bilayer. Subcellular structures, including clathrin-coated pits, caveolae, filopodia, lamellipodia, podosomes, and other intracellular membrane systems, are molded into their specific submicron-scale shapes through various mechanisms. Cells construct their micro-structures on plasma membrane and execute vital functions for life, such as cell migration, cell division, endocytosis, exocytosis, and cytoskeletal regulation. The plasma membrane, rich in anionic phospholipids, utilizes the electrostatic nature of the lipids, specifically the phosphoinositides, to form interactions with cytosolic proteins. These cytosolic proteins have three modes of interaction: 1) electrostatic interaction through unstructured polycationic regions, 2) through structured phosphoinositide-specific binding domains, and 3) through structured domains that bind the membrane without specificity for particular phospholipid. Among the structured domains, there are several that have membrane-deforming activity, which is essential for the formation of concave or convex membrane curvature. These domains include the amphipathic helix, which deforms the membrane by hemi-insertion of the helix with both hydrophobic and electrostatic interactions, and/or the BAR domain superfamily, known to use their positively charged, curved structural surface to deform membranes. Below the membrane, actin filaments support the micro-structures through interactions with several BAR proteins as well as other scaffold proteins, resulting in outward and inward membrane micro-structure formation. Here, we describe the characteristics of phospholipids, and the mechanisms utilized by phosphoinositides to regulate cellular events. We then summarize the precise mechanisms underlying the construction of membrane micro-structures and their involvements in physiological and pathological processes.
Collapse
Affiliation(s)
- Shiro Suetsugu
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, Japan; and Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Shusaku Kurisu
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, Japan; and Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Tadaomi Takenawa
- Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Nara, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, Japan; and Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| |
Collapse
|
16
|
CIP4 promotes lung adenocarcinoma metastasis and is associated with poor prognosis. Oncogene 2014; 34:3527-35. [PMID: 25174397 PMCID: PMC4978543 DOI: 10.1038/onc.2014.280] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 12/28/2022]
Abstract
Aberrant Epidermal growth factor receptor (EGFR) signaling in non-small cell lung cancer (NSCLC) is linked to tumor progression, metastasis, and poor survival rates. Here, we report the role of Cdc42-interacting protein 4 (CIP4) in the regulation of NSCLC cell invasiveness and tumor metastasis. CIP4 was highly expressed in a panel of NSCLC cell lines and normal lung epithelial cell lines. Stable knock-down (KD) of CIP4 in lung adenocarcinoma H1299 cells, expressing wild-type EGFR, led to increased EGFR levels on the cell surface, and defects in sustained activation of Erk kinase in H1299 cells treated with EGF. CIP4 localized to leading edge projections in NSCLC cells, and CIP4 KD cells displayed defects in EGF-induced cell motility and invasion through extracellular matrix. This correlated with reduced expression and activity of matrix metalloproteinase-2 (MMP-2) in CIP4 KD cells compared to control. In xenograft assays, CIP4 silencing had no effect on tumor growth, but resulted in significant defects in spontaneous metastases to the lungs from these subcutaneous tumors. This correlated with reduced expression of the Erk target gene Zeb1, and the Zeb1 target gene MMP-2 in CIP4 KD tumors compared to control. CIP4 also enhanced rates of metastasis to the liver and lungs in an intrasplenic experimental metastasis model. In human NSCLC tumor sections, CIP4 expression was elevated ≥ 2-fold in 43% of adenocarcinomas and 32% of squamous carcinomas compared to adjacent normal lung tissues. Analysis of microarray data for NSCLC patients also revealed that high CIP4 transcript levels correlated with reduced overall survival. Together, these results identify CIP4 as a positive regulator of NSCLC metastasis, and a potential poor prognostic biomarker in lung adenocarcinoma.
Collapse
|
17
|
Cytoskeletal and signaling mechanisms of neurite formation. Cell Tissue Res 2014; 359:267-78. [PMID: 25080065 DOI: 10.1007/s00441-014-1955-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/01/2014] [Indexed: 10/25/2022]
Abstract
The formation of a neurite, the basis for axons and dendrites, begins with the concerted accumulation and organization of actin and microtubules. Whereas much is known about the proteins that play a role in these processes, because they perform similar functions in axon branching and filopodia formation, much remains to be discovered concerning the interaction of these individual cytoskeletal regulators during neurite formation. Here, we review the literature regarding various models of filopodial formation and the way in which proteins that control actin organization and polymerization induce neurite formation. Although several different regulators of actin polymerization are involved in neurite initiation, redundancy occurs between these regulators, as the effects of the loss of a single regulator can be mitigated by the addition of neurite-promoting substrates and proteins. Similar to actin dynamics, both microtubule stabilizing and destabilizing proteins play a role in neurite initiation. Furthermore, interactions between the actin and microtubule cytoskeleton are required for neurite formation. Several lines of evidence indicate that the interactions between these two components of the cytoskeleton are needed for force generation and for the localization of microtubules at sites of nascent neurites. The general theme that emerges is the existence of several central regulatory pathways on which extracellular cues converge to control and organize both actin and microtubules to induce the formation of neurites.
Collapse
|
18
|
Cellular control of cortical actin nucleation. Curr Biol 2014; 24:1628-1635. [PMID: 25017211 PMCID: PMC4110400 DOI: 10.1016/j.cub.2014.05.069] [Citation(s) in RCA: 178] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/02/2014] [Accepted: 05/28/2014] [Indexed: 11/20/2022]
Abstract
The contractile actin cortex is a thin layer of actin, myosin, and actin-binding proteins that subtends the membrane of animal cells. The cortex is the main determinant of cell shape and plays a fundamental role in cell division [1-3], migration [4], and tissue morphogenesis [5]. For example, cortex contractility plays a crucial role in amoeboid migration of metastatic cells [6] and during division, where its misregulation can lead to aneuploidy [7]. Despite its importance, our knowledge of the cortex is poor, and even the proteins nucleating it remain unknown, though a number of candidates have been proposed based on indirect evidence [8-15]. Here, we used two independent approaches to identify cortical actin nucleators: a proteomic analysis using cortex-rich isolated blebs, and a localization/small hairpin RNA (shRNA) screen searching for phenotypes with a weakened cortex or altered contractility. This unbiased study revealed that two proteins generated the majority of cortical actin: the formin mDia1 and the Arp2/3 complex. Each nucleator contributed a similar amount of F-actin to the cortex but had very different accumulation kinetics. Electron microscopy examination revealed that each nucleator affected cortical network architecture differently. mDia1 depletion led to failure in division, but Arp2/3 depletion did not. Interestingly, despite not affecting division on its own, Arp2/3 inhibition potentiated the effect of mDia1 depletion. Our findings indicate that the bulk of the actin cortex is nucleated by mDia1 and Arp2/3 and suggest a mechanism for rapid fine-tuning of cortex structure and mechanics by adjusting the relative contribution of each nucleator.
Collapse
|
19
|
Kotak S, Busso C, Gönczy P. NuMA interacts with phosphoinositides and links the mitotic spindle with the plasma membrane. EMBO J 2014; 33:1815-30. [PMID: 24996901 DOI: 10.15252/embj.201488147] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The positioning and the elongation of the mitotic spindle must be carefully regulated. In human cells, the evolutionary conserved proteins LGN/Gαi1-3 anchor the coiled-coil protein NuMA and dynein to the cell cortex during metaphase, thus ensuring proper spindle positioning. The mechanisms governing cortical localization of NuMA and dynein during anaphase remain more elusive. Here, we report that LGN/Gαi1-3 are dispensable for NuMA-dependent cortical dynein enrichment during anaphase. We further establish that NuMA is excluded from the equatorial region of the cell cortex in a manner that depends on the centralspindlin components CYK4 and MKLP1. Importantly, we reveal that NuMA can directly associate with PtdInsP (PIP) and PtdInsP2 (PIP2) phosphoinositides in vitro. Furthermore, chemical or enzymatic depletion of PIP/PIP2 prevents NuMA cortical localization during mitosis, and conversely, increasing PIP2 levels augments mitotic cortical NuMA. Overall, our study uncovers a novel function for plasma membrane phospholipids in governing cortical NuMA distribution and thus the proper execution of mitosis.
Collapse
Affiliation(s)
- Sachin Kotak
- School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Coralie Busso
- School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Pierre Gönczy
- School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| |
Collapse
|
20
|
Spracklen AJ, Fagan TN, Lovander KE, Tootle TL. The pros and cons of common actin labeling tools for visualizing actin dynamics during Drosophila oogenesis. Dev Biol 2014; 393:209-226. [PMID: 24995797 DOI: 10.1016/j.ydbio.2014.06.022] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/04/2014] [Accepted: 06/17/2014] [Indexed: 10/25/2022]
Abstract
Dynamic remodeling of the actin cytoskeleton is required for both development and tissue homeostasis. While fixed image analysis has provided significant insight into such events, a complete understanding of cytoskeletal dynamics requires live imaging. Numerous tools for the live imaging of actin have been generated by fusing the actin-binding domain from an actin-interacting protein to a fluorescent protein. Here we comparatively assess the utility of three such tools--Utrophin, Lifeact, and F-tractin--for characterizing the actin remodeling events occurring within the germline-derived nurse cells during Drosophila mid-oogenesis or follicle development. Specifically, we used the UAS/GAL4 system to express these tools at different levels and in different cells, and analyzed these tools for effects on fertility, alterations in the actin cytoskeleton, and ability to label filamentous actin (F-actin) structures by both fixed and live imaging. While both Utrophin and Lifeact robustly label F-actin structures within the Drosophila germline, when strongly expressed they cause sterility and severe actin defects including cortical actin breakdown resulting in multi-nucleate nurse cells, early F-actin filament and aggregate formation during stage 9 (S9), and disorganized parallel actin filament bundles during stage 10B (S10B). However, by using a weaker germline GAL4 driver in combination with a higher temperature, Utrophin can label F-actin with minimal defects. Additionally, strong Utrophin expression within the germline causes F-actin formation in the nurse cell nuclei and germinal vesicle during mid-oogenesis. Similarly, Lifeact expression results in nuclear F-actin only within the germinal vesicle. F-tractin expresses at a lower level than the other two labeling tools, but labels cytoplasmic F-actin structures well without causing sterility or striking actin defects. Together these studies reveal how critical it is to evaluate the utility of each actin labeling tool within the tissue and cell type of interest in order to identify the tool that represents the best compromise between acceptable labeling and minimal disruption of the phenomenon being observed. In this case, we find that F-tractin, and perhaps Utrophin, when Utrophin expression levels are optimized to label efficiently without causing actin defects, can be used to study F-actin dynamics within the Drosophila nurse cells.
Collapse
Affiliation(s)
- Andrew J Spracklen
- Anatomy and Cell Biology Department, Carver College of Medicine, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Tiffany N Fagan
- Anatomy and Cell Biology Department, Carver College of Medicine, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Kaylee E Lovander
- Anatomy and Cell Biology Department, Carver College of Medicine, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Tina L Tootle
- Anatomy and Cell Biology Department, Carver College of Medicine, University of Iowa, 51 Newton Rd, Iowa City, IA 52242, USA.
| |
Collapse
|
21
|
CIP4 is required for the hypertrophic growth of neonatal cardiac myocytes. J Biomed Sci 2013; 20:56. [PMID: 23915320 PMCID: PMC3750294 DOI: 10.1186/1423-0127-20-56] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 08/01/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CIP4 is a scaffold protein that regulates membrane deformation and tubulation, organization of the actin cytoskeleton, endocytosis of growth factor receptors, and vesicle trafficking. Although expressed in the heart, CIP4 has not been studied with regards to its potential function in cardiac myocytes. RESULTS We now show using RNA interference that CIP4 expression in neonatal rat ventricular myocytes is required for the induction of non-mitotic, hypertrophic growth by the α-adrenergic agonist phenylephrine, the IL-6 cytokine leukemia inhibitor factor, and fetal bovine serum, as assayed using morphometry, immunocytochemistry for the hypertrophic marker atrial natriuretic factor and [3H]leucine incorporation for de novo protein synthesis. This requirement was consistent with the induction of CIP4 expression by hypertrophic stimulation. The inhibition of myocyte hypertrophy by CIP4 small interfering oligonucleotides (siRNA) was rescued by expression of a recombinant CIP4 protein, but not by a mutant lacking the N-terminal FCH domain responsible for CIP4 intracellular localization. CONCLUSIONS These results imply that CIP4 plays a significant role in the intracellular hypertrophic signal transduction network that controls the growth of cardiac myocytes in heart disease.
Collapse
|