1
|
Becchetti A. Interplay of Ca 2+ and K + signals in cell physiology and cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:15-46. [PMID: 38007266 DOI: 10.1016/bs.ctm.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
The cytoplasmic Ca2+ concentration and the activity of K+ channels on the plasma membrane regulate cellular processes ranging from mitosis to oriented migration. The interplay between Ca2+ and K+ signals is intricate, and different cell types rely on peculiar cellular mechanisms. Derangement of these mechanisms accompanies the neoplastic progression. The calcium signals modulated by voltage-gated (KV) and calcium-dependent (KCa) K+ channel activity regulate progression of the cell division cycle, the release of growth factors, apoptosis, cell motility and migration. Moreover, KV channels regulate the cell response to the local microenvironment by assembling with cell adhesion and growth factor receptors. This chapter summarizes the pathophysiological roles of Ca2+ and K+ fluxes in normal and cancer cells, by concentrating on several biological systems in which these functions have been studied in depth, such as early embryos, mammalian cell lines, T lymphocytes, gliomas and colorectal cancer cells. A full understanding of the underlying mechanisms will offer a comprehensive view of the ion channel implication in cancer biology and suggest potential pharmacological targets for novel therapeutic approaches in oncology.
Collapse
Affiliation(s)
- Andrea Becchetti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy.
| |
Collapse
|
2
|
Manoharan GB, Laurini C, Bottone S, Ben Fredj N, Abankwa DK. K-Ras Binds Calmodulin-Related Centrin1 with Potential Implications for K-Ras Driven Cancer Cell Stemness. Cancers (Basel) 2023; 15:3087. [PMID: 37370697 DOI: 10.3390/cancers15123087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Recent data suggest that K-Ras4B (hereafter K-Ras) can drive cancer cell stemness via calmodulin (CaM)-dependent, non-canonical Wnt-signalling. Here we examined whether another Ca2+-binding protein, the CaM-related centrin1, binds to K-Ras and could mediate some K-Ras functions that were previously ascribed to CaM. While CaM and centrin1 appear to distinguish between peptides that were derived from their classical targets, they both bind to K-Ras in cells. Cellular BRET- and immunoprecipitation data suggest that CaM engages more with K-Ras than centrin1 and that the interaction with the C-terminal membrane anchor of K-Ras is sufficient for this. Surprisingly, binding of neither K-Ras nor its membrane anchor alone to CaM or centrin1 is sensitive to inhibition of prenylation. In support of an involvement of the G-domain of K-Ras in cellular complexes with these Ca2+-binding proteins, we find that oncogenic K-RasG12V displays increased engagement with both CaM and centrin1. This is abrogated by addition of the D38A effector-site mutation, suggesting that K-RasG12V is held together with CaM or centrin1 in complexes with effectors. When treated with CaM inhibitors, the BRET-interaction of K-RasG12V with centrin1 was also disrupted in the low micromolar range, comparable to that with CaM. While CaM predominates in regulating functional membrane anchorage of K-Ras, it has a very similar co-distribution with centrin1 on mitotic organelles. Given these results, a significant overlap of the CaM- and centrin1-dependent functions of K-Ras is suggested.
Collapse
Affiliation(s)
- Ganesh Babu Manoharan
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Christina Laurini
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Sara Bottone
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Nesrine Ben Fredj
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Daniel Kwaku Abankwa
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
3
|
Abstract
The transformation of a single fertilised egg into an adult human consisting of tens of trillions of highly diverse cell types is a marvel of biology. The expansion is largely achieved by cell duplication through the process of mitosis. Mitosis is essential for normal growth, development, and tissue repair and is one of the most tightly regulated biological processes studied. This regulation is designed to ensure accurate segregation of chromosomes into each new daughter cell since errors in this process can lead to genetic imbalances, aneuploidy, that can lead to diseases including cancer. Understanding how mitosis operates and the molecular mechanisms that ensure its fidelity are therefore not only of significant intellectual value but provide unique insights into disease pathology. The purpose of this review is to revisit historical evidence that mitosis can be influenced by the ubiquitous second messenger calcium and to discuss this in the context of new findings revealing exciting new information about its role in cell division.
Collapse
Affiliation(s)
- Charlotte Nugues
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Nordine Helassa
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lee P. Haynes
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
4
|
Yousefi R, Jevdokimenko K, Kluever V, Pacheu-Grau D, Fornasiero EF. Influence of Subcellular Localization and Functional State on Protein Turnover. Cells 2021; 10:cells10071747. [PMID: 34359917 PMCID: PMC8306977 DOI: 10.3390/cells10071747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 12/14/2022] Open
Abstract
Protein homeostasis is an equilibrium of paramount importance that maintains cellular performance by preserving an efficient proteome. This equilibrium avoids the accumulation of potentially toxic proteins, which could lead to cellular stress and death. While the regulators of proteostasis are the machineries controlling protein production, folding and degradation, several other factors can influence this process. Here, we have considered two factors influencing protein turnover: the subcellular localization of a protein and its functional state. For this purpose, we used an imaging approach based on the pulse-labeling of 17 representative SNAP-tag constructs for measuring protein lifetimes. With this approach, we obtained precise measurements of protein turnover rates in several subcellular compartments. We also tested a selection of mutants modulating the function of three extensively studied proteins, the Ca2+ sensor calmodulin, the small GTPase Rab5a and the brain creatine kinase (CKB). Finally, we followed up on the increased lifetime observed for the constitutively active Rab5a (Q79L), and we found that its stabilization correlates with enlarged endosomes and increased interaction with membranes. Overall, our data reveal that both changes in protein localization and functional state are key modulators of protein turnover, and protein lifetime fluctuations can be considered to infer changes in cellular behavior.
Collapse
Affiliation(s)
- Roya Yousefi
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany;
| | - Kristina Jevdokimenko
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
| | - Verena Kluever
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
| | - David Pacheu-Grau
- Department of Cellular Biochemistry, University Medical Center Göttingen, 37073 Göttingen, Germany;
| | - Eugenio F. Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; (R.Y.); (K.J.); (V.K.)
- Correspondence:
| |
Collapse
|
5
|
Okutachi S, Manoharan GB, Kiriazis A, Laurini C, Catillon M, McCormick F, Yli-Kauhaluoma J, Abankwa D. A Covalent Calmodulin Inhibitor as a Tool to Study Cellular Mechanisms of K-Ras-Driven Stemness. Front Cell Dev Biol 2021; 9:665673. [PMID: 34307350 PMCID: PMC8296985 DOI: 10.3389/fcell.2021.665673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/04/2021] [Indexed: 11/23/2022] Open
Abstract
Recently, the highly mutated oncoprotein K-Ras4B (hereafter K-Ras) was shown to drive cancer cell stemness in conjunction with calmodulin (CaM). We previously showed that the covalent CaM inhibitor ophiobolin A (OphA) can potently inhibit K-Ras stemness activity. However, OphA, a fungus-derived natural product, exhibits an unspecific, broad toxicity across all phyla. Here we identified a less toxic, functional analog of OphA that can efficiently inactivate CaM by covalent inhibition. We analyzed a small series of benzazulenones, which bear some structural similarity to OphA and can be synthesized in only six steps. We identified the formyl aminobenzazulenone 1, here named Calmirasone1, as a novel and potent covalent CaM inhibitor. Calmirasone1 has a 4-fold increased affinity for CaM as compared to OphA and was active against K-Ras in cells within minutes, as compared to hours required by OphA. Calmirasone1 displayed a 2.5–4.5-fold higher selectivity for KRAS over BRAF mutant 3D spheroid growth than OphA, suggesting improved relative on-target activity. Importantly, Calmirasone1 has a 40–260-fold lower unspecific toxic effect on HRAS mutant cells, while it reaches almost 50% of the activity of novel K-RasG12C specific inhibitors in 3D spheroid assays. Our results suggest that Calmirasone1 can serve as a new tool compound to further investigate the cancer cell biology of the K-Ras and CaM associated stemness activities.
Collapse
Affiliation(s)
- Sunday Okutachi
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ganesh Babu Manoharan
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexandros Kiriazis
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Christina Laurini
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marie Catillon
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Frank McCormick
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States.,Frederick National Laboratory for Cancer Research, Cancer Research Technology Program, Leidos Biomedical Research, Inc., National Cancer Institute RAS Initiative, Frederick, MD, United States
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Daniel Abankwa
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
6
|
Promotion of cancer cell stemness by Ras. Biochem Soc Trans 2021; 49:467-476. [PMID: 33544116 PMCID: PMC7925005 DOI: 10.1042/bst20200964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/17/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSC) may be the most relevant and elusive cancer cell population, as they have the exquisite ability to seed new tumors. It is plausible, that highly mutated cancer genes, such as KRAS, are functionally associated with processes contributing to the emergence of stemness traits. In this review, we will summarize the evidence for a stemness driving activity of oncogenic Ras. This activity appears to differ by Ras isoform, with the highly mutated KRAS having a particularly profound impact. Next to established stemness pathways such as Wnt and Hedgehog (Hh), the precise, cell cycle dependent orchestration of the MAPK-pathway appears to relay Ras activation in this context. We will examine how non-canonical activities of K-Ras4B (hereafter K-Ras) could be enabled by its trafficking chaperones calmodulin and PDE6D/PDEδ. Both dynamically localize to the cellular machinery that is intimately linked to cell fate decisions, such as the primary cilium and the centrosome. Thus, it can be speculated that oncogenic K-Ras disrupts fundamental polarized signaling and asymmetric apportioning processes that are necessary during cell differentiation.
Collapse
|
7
|
Zhu G. The Biophysical Society of Hong Kong (BPHK): past, present, and future. Biophys Rev 2019; 11:259-261. [PMID: 31055758 DOI: 10.1007/s12551-019-00525-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 04/15/2019] [Indexed: 11/27/2022] Open
Affiliation(s)
- Guang Zhu
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
8
|
Ayad O, Magaud C, Sebille S, Bescond J, Mimbimi C, Cognard C, Faivre JF, Bois P, Chatelier A. Functional BKCa channel in human resident cardiac stem cells expressing W8B2. FEBS J 2017; 285:518-530. [PMID: 29211342 DOI: 10.1111/febs.14352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 11/02/2017] [Accepted: 11/30/2017] [Indexed: 01/24/2023]
Abstract
Recently, a new population of resident cardiac stem cells (CSCs) positive for the W8B2 marker has been identified. These CSCs are considered to be an ideal cellular source to repair myocardial damage after infarction. However, the electrophysiological profile of these cells has not been characterized yet. We first establish the conditions of isolation and expansion of W8B2+ CSCs from human heart biopsies using a magnetic sorting system followed by flow cytometry cell sorting. These cells display a spindle-shaped morphology, are highly proliferative, and possess self-renewal capacity demonstrated by their ability to form colonies. Besides, W8B2+ CSCs are positive for mesenchymal markers but negative for hematopoietic and endothelial ones. RT-qPCR and immunostaining experiments show that W8B2+ CSCs express some early cardiac-specific transcription factors but lack the expression of cardiac-specific structural genes. Using patch clamp in the whole-cell configuration, we show for the first time the electrophysiological signature of BKCa current in these cells. Accordingly, RT-PCR and western blotting analysis confirmed the presence of BKCa at both mRNA and protein levels in W8B2+ CSCs. Interestingly, BKCa channel inhibition by paxilline decreased cell proliferation in a concentration-dependent manner and halted cell cycle progression at the G0/G1 phase. The inhibition of BKCa also decreased the self-renewal capacity but did not affect migration of W8B2+ CSCs. Taken together, our results are consistent with an important role of BKCa channels in cell cycle progression and self-renewal in human cardiac stem cells.
Collapse
Affiliation(s)
- Oualid Ayad
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Christophe Magaud
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Stéphane Sebille
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jocelyn Bescond
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Chloé Mimbimi
- Service de chirurgie cardio-thoracique, CHU Poitiers, France
| | - Christian Cognard
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Jean-Francois Faivre
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Patrick Bois
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| | - Aurelien Chatelier
- Equipe Transferts Ioniques et Rythmicité Cardiaque, Laboratoire Signalisation et Transports Ioniques Membranaires, CNRS ERL 7368, EA 7349, Université de Poitiers, France
| |
Collapse
|
9
|
Calcium signaling and cell cycle: Progression or death. Cell Calcium 2017; 70:3-15. [PMID: 28801101 DOI: 10.1016/j.ceca.2017.07.006] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/23/2017] [Accepted: 07/23/2017] [Indexed: 12/12/2022]
Abstract
Cytosolic Ca2+ concentration levels fluctuate in an ordered manner along the cell cycle, in line with the fact that Ca2+ is involved in the regulation of cell proliferation. Cell proliferation should be an error-free process, yet is endangered by mistakes. In fact, a complex network of proteins ensures that cell cycle does not progress until the previous phase has been successfully completed. Occasionally, errors occur during the cell cycle leading to cell cycle arrest. If the error is severe, and the cell cycle checkpoints work perfectly, this results into cellular demise by activation of apoptotic or non-apoptotic cell death programs. Cancer is characterized by deregulated proliferation and resistance against cell death. Ca2+ is a central key to these phenomena as it modulates signaling pathways that control oncogenesis and cancer progression. Here, we discuss how Ca2+ participates in the exogenous and endogenous signals controlling cell proliferation, as well as in the mechanisms by which cells die if irreparable cell cycle damage occurs. Moreover, we summarize how Ca2+ homeostasis remodeling observed in cancer cells contributes to deregulated cell proliferation and resistance to cell death. Finally, we discuss the possibility to target specific components of Ca2+ signal pathways to obtain cytostatic or cytotoxic effects.
Collapse
|
10
|
Verma D, Sakuntala M, Murmu A, Bhattacharya A, Chary KVR. 1H, 13C and 15N NMR assignments of an unusual Ca 2+-binding protein from Entamoeba histolytica in its apo form. BIOMOLECULAR NMR ASSIGNMENTS 2017; 11:63-67. [PMID: 27990612 DOI: 10.1007/s12104-016-9721-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 12/01/2016] [Indexed: 06/06/2023]
Abstract
We report almost complete sequence specific 1H, 13C and 15N NMR assignments of an unusual Ca2+-binding protein from Entamoeba histolytica (EhCaBP6) in its apo form as a prelude to its structural and functional characterization.
Collapse
Affiliation(s)
- Deepshikha Verma
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Colaba, Mumbai, 400005, India
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, 21, Brindavan Colony, Narsingi, Hyderabad, 500075, India
| | - Mutyala Sakuntala
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Colaba, Mumbai, 400005, India
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, 21, Brindavan Colony, Narsingi, Hyderabad, 500075, India
| | - Aruna Murmu
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Alok Bhattacharya
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Kandala V R Chary
- Department of Chemical Sciences, Tata Institute of Fundamental Research, 1, Homi Bhabha Road, Colaba, Mumbai, 400005, India.
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, 21, Brindavan Colony, Narsingi, Hyderabad, 500075, India.
| |
Collapse
|
11
|
Ca 2+ Signalling and Membrane Dynamics During Cytokinesis in Animal Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 981:389-412. [PMID: 29594869 DOI: 10.1007/978-3-319-55858-5_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
Interest in the role of Ca2+ signalling as a possible regulator of the combinatorial processes that result in the separation of the daughter cells during cytokinesis, extend back almost a 100 years. One of the key processes required for the successful completion of cytokinesis in animal cells (especially in the large holoblastically and meroblastically dividing embryonic cells of a number of amphibian and fish species), is the dynamic remodelling of the plasma membrane. Ca2+ signalling was subsequently demonstrated to regulate various different aspects of cytokinesis in animal cells, and so here we focus specifically on the role of Ca2+ signalling in the remodelling of the plasma membrane. We begin by providing a brief history of the animal models used and the research accomplished by the early twentieth century investigators, with regards to this aspect of animal cell cytokinesis. We then review the most recent progress made (i.e., in the last 10 years), which has significantly advanced our current understanding on the role of cytokinetic Ca2+ signalling in membrane remodelling. To this end, we initially summarize what is currently known about the Ca2+ transients generated during animal cell cytokinesis, and then we describe the latest findings regarding the source of Ca2+ generating these transients. Finally, we review the current evidence about the possible targets of the different cytokinetic Ca2+ transients with a particular emphasis on those that either directly or indirectly affect plasma membrane dynamics. With regards to the latter, we discuss the possible role of the early Ca2+ signalling events in the deformation of the plasma membrane at the start of cytokinesis (i.e., during furrow positioning), as well as the role of the subsequent Ca2+ signals in the trafficking and fusion of vesicles, which help to remodel the plasma membrane during the final stages of cell division. As it is becoming clear that each of the cytokinetic Ca2+ transients might have multiple, integrated targets, deciphering the precise role of each transient represents a significant (and ongoing) challenge.
Collapse
|
12
|
Li R, Leblanc J, He K, Liu XJ. Spindle function in Xenopus oocytes involves possible nanodomain calcium signaling. Mol Biol Cell 2016; 27:3273-3283. [PMID: 27582389 PMCID: PMC5170860 DOI: 10.1091/mbc.e16-05-0338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/25/2016] [Indexed: 11/11/2022] Open
Abstract
Injection of dibromo-BAPTA caused immediate collapse of meiotic spindles in frog oocytes. In contrast, EGTA had no effect on the spindle or polar body emission. The disruption of spindle integrity by the fast but not slow calcium chelators suggests that meiotic spindle function in the oocytes involves nanodomain calcium signaling. Intracellular calcium transients are a universal phenomenon at fertilization and are required for egg activation, but the exact role of Ca2+ in second-polar-body emission remains unknown. On the other hand, similar calcium transients have not been demonstrated during oocyte maturation, and yet, manipulating intracellular calcium levels interferes with first-polar-body emission in mice and frogs. To determine the precise role of calcium signaling in polar body formation, we used live-cell imaging coupled with temporally precise intracellular calcium buffering. We found that BAPTA-based calcium chelators cause immediate depolymerization of spindle microtubules in meiosis I and meiosis II. Surprisingly, EGTA at similar or higher intracellular concentrations had no effect on spindle function or polar body emission. Using two calcium probes containing permutated GFP and the calcium sensor calmodulin (Lck-GCaMP3 and GCaMP3), we demonstrated enrichment of the probes at the spindle but failed to detect calcium increase during oocyte maturation at the spindle or elsewhere. Finally, endogenous calmodulin was found to colocalize with spindle microtubules throughout all stages of meiosis. Our results—most important, the different sensitivities of the spindle to BAPTA and EGTA—suggest that meiotic spindle function in frog oocytes requires highly localized, or nanodomain, calcium signaling.
Collapse
Affiliation(s)
- Ruizhen Li
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada
| | - Julie Leblanc
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kevin He
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada
| | - X Johné Liu
- Ottawa Hospital Research Institute, Ottawa Hospital-General Campus, Ottawa, ON K1H 8L6, Canada .,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada.,Department of Obstetrics and Gynaecology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
13
|
Zhou Q, Hu H, Li Z. An EF-hand-containing Protein in Trypanosoma brucei Regulates Cytokinesis Initiation by Maintaining the Stability of the Cytokinesis Initiation Factor CIF1. J Biol Chem 2016; 291:14395-409. [PMID: 27226595 DOI: 10.1074/jbc.m116.726133] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Indexed: 11/06/2022] Open
Abstract
Trypanosoma brucei undergoes cytokinesis uni-directionally from the anterior tip of the new flagellum attachment zone (FAZ) toward the posterior end of the cell. We recently delineated a novel signaling pathway composed of polo-like kinase, cytokinesis initiation factor 1 (CIF1), and aurora B kinase that acts in concert at the new FAZ tip to regulate cytokinesis initiation. To identify new cytokinesis regulators, we carried out proximity-dependent biotin identification and identified many CIF1 binding partners and near neighbors. Here we report a novel CIF1-binding protein, named CIF2, and its mechanistic role in cytokinesis initiation. CIF2 interacts with CIF1 in vivo and co-localizes with CIF1 at the new FAZ tip during early cell cycle stages. RNAi of CIF2 inhibited the normal, anterior-to-posterior cytokinesis but activated an alternative, posterior-to-anterior cytokinesis. CIF2 depletion destabilized CIF1 and disrupted the localization of polo-like kinase and aurora B kinase to the new FAZ tip, thus revealing the mechanistic role of CIF2 in cytokinesis initiation. Surprisingly, overexpression of CIF2 also inhibited the normal, anterior-to-posterior cytokinesis and triggered the alternative, posterior-to-anterior cytokinesis, suggesting a tight control of CIF2 protein abundance. These results identified a new regulator in the cytokinesis regulatory pathway and reiterated that a backup cytokinesis pathway is activated by inhibiting the normal cytokinesis pathway.
Collapse
Affiliation(s)
- Qing Zhou
- From the Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Huiqing Hu
- From the Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Ziyin Li
- From the Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas 77030
| |
Collapse
|
14
|
Contractile dynamics change before morphological cues during fluorescence [corrected] illumination. Sci Rep 2015; 5:18513. [PMID: 26691776 PMCID: PMC4686977 DOI: 10.1038/srep18513] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/19/2015] [Indexed: 01/15/2023] Open
Abstract
Illumination can have adverse effects on live cells. However, many experiments, e.g. traction force microscopy, rely on fluorescence microscopy. Current methods to assess undesired photo-induced cell changes rely on qualitative observation of changes in cell morphology. Here we utilize a quantitative technique to identify the effect of light on cell contractility prior to morphological changes. Fibroblasts were cultured on soft elastic hydrogels embedded with fluorescent beads. The adherent cells generated contractile forces that deform the substrate. Beads were used as fiducial markers to quantify the substrate deformation over time, which serves as a measure of cell force dynamics. We find that cells exposed to moderate fluorescence illumination (λ = 540–585 nm, I = 12.5 W/m2, duration = 60 s) exhibit rapid force relaxation. Strikingly, cells exhibit force relaxation after only 2 s of exposure, suggesting that photo-induced relaxation occurs nearly immediately. Evidence of photo-induced morphological changes were not observed for 15–30 min after illumination. Force relaxation and morphological changes were found to depend on wavelength and intensity of excitation light. This study demonstrates that changes in cell contractility reveal evidence of a photo-induced cell response long before any morphological cues.
Collapse
|
15
|
A calmodulin like EF hand protein positively regulates oxalate decarboxylase expression by interacting with E-box elements of the promoter. Sci Rep 2015; 5:14578. [PMID: 26455820 PMCID: PMC4600981 DOI: 10.1038/srep14578] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/03/2015] [Indexed: 12/02/2022] Open
Abstract
Oxalate decarboxylase (OXDC) enzyme has immense biotechnological applications due to its ability to decompose anti-nutrient oxalic acid. Flammulina velutipes, an edible wood rotting fungus responds to oxalic acid by induction of OXDC to maintain steady levels of pH and oxalate anions outside the fungal hyphae. Here, we report that upon oxalic acid induction, a calmodulin (CaM) like protein-FvCaMLP, interacts with the OXDC promoter to regulate its expression. Electrophoretic mobility shift assay showed that FvCamlp specifically binds to two non-canonical E-box elements (AACGTG) in the OXDC promoter. Moreover, substitutions of amino acids in the EF hand motifs resulted in loss of DNA binding ability of FvCamlp. F. velutipes mycelia treated with synthetic siRNAs designed against FvCaMLP showed significant reduction in FvCaMLP as well as OXDC transcript pointing towards positive nature of the regulation. FvCaMLP is different from other known EF hand proteins. It shows sequence similarity to both CaMs and myosin regulatory light chain (Cdc4), but has properties typical of a calmodulin, like binding of 45Ca2+, heat stability and Ca2+ dependent electrophoretic shift. Hence, FvCaMLP can be considered a new addition to the category of unconventional Ca2+ binding transcriptional regulators.
Collapse
|
16
|
Kulkarni C, Lo M, Fraseur JG, Tirrell DA, Kinzer-Ursem TL. Bioorthogonal Chemoenzymatic Functionalization of Calmodulin for Bioconjugation Applications. Bioconjug Chem 2015; 26:2153-60. [PMID: 26431265 DOI: 10.1021/acs.bioconjchem.5b00449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Calmodulin (CaM) is a widely studied Ca(2+)-binding protein that is highly conserved across species and involved in many biological processes, including vesicle release, cell proliferation, and apoptosis. To facilitate biophysical studies of CaM, researchers have tagged and mutated CaM at various sites, enabling its conjugation to fluorophores, microarrays, and other reactive partners. However, previous attempts to add a reactive label to CaM for downstream studies have generally employed nonselective labeling methods or resulted in diminished CaM function. Here we report the first engineered CaM protein that undergoes site-specific and bioorthogonal labeling while retaining wild-type activity levels. By employing a chemoenzymatic labeling approach, we achieved selective and quantitative labeling of the engineered CaM protein with an N-terminal 12-azidododecanoic acid tag; notably, addition of the tag did not interfere with the ability of CaM to bind Ca(2+) or a partner protein. The specificity of our chemoenzymatic labeling approach also allowed for selective conjugation of CaM to reactive partners in bacterial cell lysates, without intermediate purification of the engineered protein. Additionally, we prepared CaM-affinity resins that were highly effective in purifying a representative CaM-binding protein, demonstrating that the engineered CaM remains active even after surface capture. Beyond studies of CaM and CaM-binding proteins, the protein engineering and surface capture methods described here should be translatable to other proteins and other bioconjugation applications.
Collapse
Affiliation(s)
- Chethana Kulkarni
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Megan Lo
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| | - David A Tirrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States
| | - Tamara L Kinzer-Ursem
- Division of Chemistry and Chemical Engineering, California Institute of Technology , 1200 East California Blvd., Pasadena, California 91125, United States.,Weldon School of Biomedical Engineering, Purdue University , 206 South Martin Jischke Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
17
|
Berchtold MW, Villalobo A. The many faces of calmodulin in cell proliferation, programmed cell death, autophagy, and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1843:398-435. [PMID: 24188867 DOI: 10.1016/j.bbamcr.2013.10.021] [Citation(s) in RCA: 236] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 10/24/2013] [Accepted: 10/26/2013] [Indexed: 12/21/2022]
Abstract
Calmodulin (CaM) is a ubiquitous Ca(2+) receptor protein mediating a large number of signaling processes in all eukaryotic cells. CaM plays a central role in regulating a myriad of cellular functions via interaction with multiple target proteins. This review focuses on the action of CaM and CaM-dependent signaling systems in the control of vertebrate cell proliferation, programmed cell death and autophagy. The significance of CaM and interconnected CaM-regulated systems for the physiology of cancer cells including tumor stem cells, and processes required for tumor progression such as growth, tumor-associated angiogenesis and metastasis are highlighted. Furthermore, the potential targeting of CaM-dependent signaling processes for therapeutic use is discussed.
Collapse
Key Words
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-ethyl]-4,5-dihydro-pyrazol-1-yl]-benzoic acid
- (4-[3,5-bis-[2-(4-hydroxy-3-methoxy-phenyl)-vinyl]-4,5-dihydro-pyrazol-1-yl]-phenyl)-(4-methyl-piperazin-1-yl)-methanone
- (−) enantiomer of dihydropyrine 3-methyl-5-3-(4,4-diphenyl-1-piperidinyl)-propyl-1,4-dihydro-2,6-dimethyl-4-(3-nitrophenyl)-piridine-3,5-dicarboxylate-hydrochloride (niguldipine)
- 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-l-tyrosyl]-4-phenylpiperazine
- 12-O-tetradecanoyl-phorbol-13-acetate
- 2-chloro-(ε-amino-Lys(75))-[6-(4-(N,N′-diethylaminophenyl)-1,3,5-triazin-4-yl]-CaM adduct
- 3′-(β-chloroethyl)-2′,4′-dioxo-3,5′-spiro-oxazolidino-4-deacetoxy-vinblastine
- 7,12-dimethylbenz[a]anthracene
- Apoptosis
- Autophagy
- B859-35
- CAPP(1)-CaM
- Ca(2+) binding protein
- Calmodulin
- Cancer biology
- Cell proliferation
- DMBA
- EBB
- FL-CaM
- FPCE
- HBC
- HBCP
- J-8
- KAR-2
- KN-62
- KN-93
- N-(4-aminobutyl)-2-naphthalenesulfonamide
- N-(4-aminobutyl)-5-chloro-2-naphthalenesulfonamide
- N-(6-aminohexyl)-1-naphthalenesulfonamide
- N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide
- N-8-aminooctyl-5-iodo-naphthalenesulfonamide
- N-[2-[N-(4-chlorocinnamyl)-N-methylaminomethyl]phenyl]-N-(2-hydroxyethyl)-4-methoxybenzenesulfonamide
- O-(4-ethoxyl-butyl)-berbamine
- RITC-CaM
- TA-CaM
- TFP
- TPA
- W-12
- W-13
- W-5
- W-7
- fluorescein-CaM adduct
- fluphenazine-N-2-chloroethane
- norchlorpromazine-CaM adduct
- rhodamine isothiocyanate-CaM adduct
- trifluoperazine
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, Copenhagen Biocenter 4-2-09 Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | - Antonio Villalobo
- Instituto de Investigaciones Biomédicas, Department of Cancer Biology, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
18
|
Dai G, Qian Y, Chen J, Meng FL, Pan FY, Shen WG, Zhang SZ, Xue B, Li CJ. Calmodulin activation of polo-like kinase 1 is required during mitotic entry. Biochem Cell Biol 2013; 91:287-94. [PMID: 24032677 DOI: 10.1139/bcb-2013-0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Polo-like kinase 1 (Plk1) is a conserved key regulator of the G2/M transition, but its upstream spatiotemporal regulators remain unknown. With the help of immunofluorescence, co-immunoprecipitation, and glutathione S-transferase (GST) pull-down assay, we found that calmodulin (CaM) is one such regulatory molecule that associates with Plk1 from G2 to metaphase. More importantly, this interaction results in considerable stimulation of Plk1 kinase activity leading to hyperphosphorylation of Cdc25C. Our results provide new insight into the role of CaM as an upstream regulator of Plk1 activation during mitotic entry.
Collapse
Affiliation(s)
- Gu Dai
- a Model Animal Research Center (MARC) and School of Medicine, Nanjing University, Nanjing 210093, China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Grewal JS, Padhan N, Aslam S, Bhattacharya A, Lohia A. The calcium binding protein EhCaBP6 is a microtubular-end binding protein inEntamoeba histolytica. Cell Microbiol 2013; 15:2020-33. [DOI: 10.1111/cmi.12167] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 06/28/2013] [Accepted: 07/08/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Jaspreet Singh Grewal
- Department of Biochemistry; Bose Institute; P-1/12, C. I. T. Scheme VII-M Kolkata 700 054 India
| | - Narendra Padhan
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Saima Aslam
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Alok Bhattacharya
- School of Life Sciences; Jawaharlal Nehru University; New Delhi India
| | - Anuradha Lohia
- Department of Biochemistry; Bose Institute; P-1/12, C. I. T. Scheme VII-M Kolkata 700 054 India
| |
Collapse
|
20
|
Biphasic assembly of the contractile apparatus during the first two cell division cycles in zebrafish embryos. ZYGOTE 2013; 22:218-28. [DOI: 10.1017/s0967199413000051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryThe large and optically clear embryos of the zebrafish provide an excellent model system in which to study the dynamic assembly of the essential contractile band components, actin and myosin, via double fluorescent labelling in combination with confocal microscopy. We report the rapid appearance (i.e. within <2 min) of a restricted arc of F-actin patches along the prospective furrow plane in a central, apical region of the blastodisc cortex. These patches then fused with each other end-to-end forming multiple actin cables, which were subsequently bundled together forming an F-actin band. During this initial assembly phase, the F-actin-based structure did not elongate laterally, but was still restricted to an arc extending ~15° either side of the blastodisc apex. This initial assembly phase was then followed by an extension phase, where additional F-actin patches were added to each end of the original arc, thus extending it out to the edges of the blastodisc. The dynamics of phosphorylated myosin light chain 2 (MLC2) recruitment to this F-actin scaffold also reflect the two-phase nature of the contractile apparatus assembly. MLC2 was not associated with the initial F-actin arc, but MLC2 clusters were recruited and assembled into the extending ends of the band. We propose that the MLC2-free central region of the contractile apparatus acts to position and then extend the cleavage furrow in the correct plane, while the actomyosin ends alone generate the force required for furrow ingression. This biphasic assembly strategy may be required to successfully divide the early cells of large embryos.
Collapse
|
21
|
Knockdown of PLC-gamma-2 and calmodulin 1 genes sensitizes human cervical adenocarcinoma cells to doxorubicin and paclitaxel. Cancer Cell Int 2012; 12:30. [PMID: 22709569 PMCID: PMC3407783 DOI: 10.1186/1475-2867-12-30] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 06/18/2012] [Indexed: 02/08/2023] Open
Abstract
Background RNA interference (RNAi) is a powerful approach in functional genomics to selectively silence messenger mRNA (mRNA) expression and can be employed to rapidly develop potential novel drugs against a complex disease like cancer. However, naked siRNA being anionic is unable to cross the anionic cell membrane through passive diffusion and therefore, delivery of siRNA remains a major hurdle to overcome before the potential of siRNA technology can fully be exploited in cancer. pH-sensitive carbonate apatite has recently been developed as an efficient tool to deliver siRNA into the mammalian cells by virtue of its high affinity interaction with the siRNA and the desirable size distribution of the resulting siRNA-apatite complex for effective cellular endocytosis. Moreover, internalized siRNA was found to escape from the endosomes in a time-dependent manner and efficiently silence gene expression. Results Here we show that carbonate apatite-mediated delivery of siRNA against PLC-gamma-2 (PLCG2) and calmodulin 1 (CALM1) genes has led to the sensitization of a human cervical cancer cell line to doxorubicin- and paclitaxel depending on the dosage of the individual drug whereas no such enhancement in cell death was observed with cisplatin irrespective of the dosage following intracellular delivery of the siRNAs. Conclusion Thus, PLCG2 and CALM1 genes are two potential targets for gene knockdown in doxorubicin and paclitaxel-based chemotherapy of cervical cancer.
Collapse
|
22
|
Vinflunine: a new vision that may translate into antiangiogenic and antimetastatic activity. Anticancer Drugs 2012; 23:1-11. [PMID: 22027536 DOI: 10.1097/cad.0b013e32834d237b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microtubules and tubulin are major dynamic and structural cellular components that play a key role in several cell functions, including division, signalling and intracellular trafficking. Normal epithelial cells have a highly structured, rigid cytoskeletal network that is compatible with cell motility. Thus, tubulin and microtubules are compelling cellular targets for chemotherapy. In fact, among anticancer agents, those that target microtubules constitute one of the most effective classes of chemotherapeutics in cancer. The list of compounds that target either tubulin or microtubules is extensive and consists of chemically unique compounds that bind to the tubulin dimers and destabilize microtubules (Vinca alkaloids) and those that bind to the microtubule polymer and stabilize microtubules (taxanes). Tumour-induced angiogenesis, the formation of new capillaries from existing blood vessels, and epithelial-mesenchymal transition are two steps that are critical for both tumour growth and metastatic spread. Three possible mechanisms of action are described with vinflunine, the new-generation Vinca alkaloid to arrive in clinical practice are as follows: it acts against tubulin and microtubules, disrupts newly formed blood vessels and seems to be able to reduce the metastatic process as shown in preclinical studies. These findings support the hypothesis that vinflunine, by blocking microtubule functions that contribute to cell shape, polarization, migration and other processes, might be responsible not only for tumour-cytostatic but also for specific antiangiogenic or antiepithelial-mesenchymal transition effects.
Collapse
|
23
|
Conway BR, Demarest KT. The Use of Biosensors to Study GPCR Function: Applications for High-Content Screening. ACTA ACUST UNITED AC 2011. [DOI: 10.3109/10606820214641] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
24
|
Papoff G, Trivieri N, Crielesi R, Ruberti F, Marsilio S, Ruberti G. FADD-calmodulin interaction: a novel player in cell cycle regulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:898-911. [PMID: 20420860 DOI: 10.1016/j.bbamcr.2010.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 03/26/2010] [Accepted: 04/12/2010] [Indexed: 11/16/2022]
Abstract
Analyses of knockout and mutant transgenic mice as well as in vitro studies demonstrated a complex role of FADD in the regulation of cell fate. FADD is involved in death receptor induced apoptosis, cell cycle progression and cell proliferation. In a search for mechanisms that might regulate FADD functions, we identified, upon the screening of a lambda-phage cDNA library, calmodulin (CaM) as a novel FADD interacting protein. CaM is a key mediator of signals by the secondary messenger calcium and it is an essential regulator of cell cycle progression and cell survival. Here, we describe the identification and characterization of two calcium dependent CaM binding sites in the alpha helices 8-9 and 10-11 of FADD. Phosphorylation of human FADD at the C-terminal serine 194, by casein kinase I alpha (CKIalpha), has been shown to regulate FADD-dependent non-apoptotic activities. Remarkably, we showed that both FADD and CaM are CKIalpha substrates and that in synchronized HeLa cells, FADD, CaM and CKIalpha co-localize at the mitotic spindle in metaphase and anaphase. Moreover, complementation experiments in Jurkat FADD-/- T cells indicated that: a) cells expressing FADD mutants in the CaM binding sites are protected from Taxol-induced G2/M cell cycle arrest; b) FADD/CaM interaction is not required for Fas receptor-mediated apoptosis although Fas and CaM might compete for binding to FADD. We suggest that the interplay of FADD, CaM and CKIalpha may have an important role in the regulation of cell fate.
Collapse
Affiliation(s)
- Giuliana Papoff
- Istituto di Biologia Cellulare, Consiglio Nazionale delle Ricerche, Campus A. Buzzati-Traverso, Monterotondo, Rome, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Atilla-Gokcumen GE, Castoreno AB, Sasse S, Eggert US. Making the cut: the chemical biology of cytokinesis. ACS Chem Biol 2010; 5:79-90. [PMID: 20014865 DOI: 10.1021/cb900256m] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Cytokinesis is the last step in the cell cycle, where daughter cells finally separate. It is precisely regulated in both time and space to ensure that each daughter cell receives an equal share of DNA and other cellular materials. Chemical biology approaches have been used very successfully to study the mechanism of cytokinesis. In this review, we discuss the use of small molecule probes to perturb cytokinesis, as well as the role naturally occurring small molecule metabolites such as lipids play during cytokinesis.
Collapse
Affiliation(s)
- G. Ekin Atilla-Gokcumen
- Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Adam B. Castoreno
- Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| | - Sofia Sasse
- Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
- Westfälische Wilhelms-Universität Münster, Germany
| | - Ulrike S. Eggert
- Dana-Farber Cancer Institute and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
26
|
Hsu STD, Blaser G, Behrens C, Cabrita LD, Dobson CM, Jackson SE. Folding study of Venus reveals a strong ion dependence of its yellow fluorescence under mildly acidic conditions. J Biol Chem 2009; 285:4859-69. [PMID: 19901033 DOI: 10.1074/jbc.m109.000695] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Venus is a yellow fluorescent protein that has been developed for its fast chromophore maturation rate and bright yellow fluorescence that is relatively insensitive to changes in pH and ion concentrations. Here, we present a detailed study of the stability and folding of Venus in the pH range from 6.0 to 8.0 using chemical denaturants and a variety of spectroscopic probes. By following hydrogen-deuterium exchange of (15)N-labeled Venus using NMR spectroscopy over 13 months, residue-specific free energies of unfolding of some highly protected amide groups have been determined. Exchange rates of less than one per year are observed for some amide groups. A super-stable core is identified for Venus and compared with that previously reported for green fluorescent protein. These results are discussed in terms of the stability and folding of fluorescent proteins. Under mildly acidic conditions, we show that Venus undergoes a drastic decrease in yellow fluorescence at relatively low concentrations of guanidinium chloride. A detailed study of this effect establishes that it is due to pH-dependent, nonspecific interactions of ions with the protein. In contrast to previous studies on enhanced green fluorescence protein variant S65T/T203Y, which showed a specific halide ion-binding site, NMR chemical shift mapping shows no evidence for specific ion binding. Instead, chemical shift perturbations are observed for many residues primarily located in both lids of the beta-barrel structure, which suggests that small scale structural rearrangements occur on increasing ionic strength under mildly acidic conditions and that these are propagated to the chromophore resulting in fluorescence quenching.
Collapse
Affiliation(s)
- Shang-Te Danny Hsu
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.
| | | | | | | | | | | |
Collapse
|
27
|
Hsu STD, Blaser G, Jackson SE. The folding, stability and conformational dynamics of beta-barrel fluorescent proteins. Chem Soc Rev 2009; 38:2951-65. [PMID: 19771338 DOI: 10.1039/b908170b] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This critical review describes our current knowledge on the folding, stability and conformational dynamics of fluorescent proteins (FPs). The biophysical studies that have led to the elucidation of many of the key features of the complex energy landscape for folding for GFP and its variants are discussed. These illustrate some important issues surrounding how the large beta-barrel structure forms, and will be of interest to the protein folding community. In addition, the review highlights the importance of some of these results for the use of FPs in in vivo applications. The results should facilitate and aid in experimental designs of in vivo applications, as well as the interpretation of in vivo experimental data. The review is therefore of interest to all those working with FPs in vivo (103 references).
Collapse
Affiliation(s)
- Shang-Te Danny Hsu
- Chemistry Department, University of Cambridge, Lensfield Road, Cambridge, UK CB2 1EW
| | | | | |
Collapse
|
28
|
Huang M, Wei JN, Peng WX, Liang J, Zhao C, Qian Y, Dai G, Yuan J, Pan FY, Xue B, Sha JH, Li CJ. The association of CaM and Hsp70 regulates S-phase arrest and apoptosis in a spatially and temporally dependent manner in human cells. Cell Stress Chaperones 2009; 14:343-53. [PMID: 18989758 PMCID: PMC2728269 DOI: 10.1007/s12192-008-0088-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2008] [Revised: 10/09/2008] [Accepted: 10/09/2008] [Indexed: 10/21/2022] Open
Abstract
The cell cycle is controlled by regulators functioning at the right time and at the right place. We have found that calmodulin (CaM) has specific distribution patterns during different cell-cycle stages. Here, we identify cell-cycle-specific binding proteins of CaM and examine their function during cell-cycle progression. We first applied immunoprecipitation methods to isolate CaM-binding proteins from cell lysates obtained at different cell-cycle phases and then identified these proteins using mass spectrometry methods. A total of 41 proteins were identified including zinc finger proteins, ribosomal proteins, and heat shock proteins operating in a Ca(2+)-dependent or independent manner. Fifteen proteins were shown to interact with CaM in a cell-phase-specific manner. The association of the selected proteins and CaM were confirmed with in vitro immunoprecipitation and immunostaining methods. One of the identified proteins, heat shock protein 70 (Hsp70), was further studied with respect to its cell-cycle-related function. In vivo fluorescence resonance energy transfer (FRET) analysis showed that the interaction of CaM and Hsp70 was found in the nucleus during the S phase. Overexpression of Hsp70 is shown to arrest cells at S phase and, thus, induce cell apoptosis. When we disrupted the CaM-Hsp70 association with HSP70 truncation without the CaM-binding domain, we found that S-phase arrest and apoptosis could be rescued. The results suggest that the spatial and temporal association of CaM and Hsp70 can regulate cell-cycle progression and cell apoptosis.
Collapse
Affiliation(s)
- Min Huang
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Jun-Ning Wei
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Wan-Xin Peng
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Juan Liang
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Chun Zhao
- Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029 China
| | - Yan Qian
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Gu Dai
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Jun Yuan
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Fei-Yan Pan
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
- School of Medicine, Nanjing University, Nanjing, 210095 China
| | - Bin Xue
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
- School of Medicine, Nanjing University, Nanjing, 210095 China
| | - Jia-Hao Sha
- Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 210029 China
| | - Chao-Jun Li
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
- Model Animal Research Center (MARC), Nanjing University, Nanjing, 210095 China
- School of Medicine, Nanjing University, Nanjing, 210095 China
| |
Collapse
|
29
|
Lin JJ, Li Y, Eppinga RD, Wang Q, Jin J. Chapter 1 Roles of Caldesmon in Cell Motility and Actin Cytoskeleton Remodeling. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 274:1-68. [DOI: 10.1016/s1937-6448(08)02001-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
30
|
Zhou L, Chang DC. Dynamics and structure of the Bax-Bak complex responsible for releasing mitochondrial proteins during apoptosis. J Cell Sci 2008; 121:2186-96. [PMID: 18544634 DOI: 10.1242/jcs.024703] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bax and Bak are known to play a central role in facilitating the release of mitochondrial intermembrane proteins during apoptosis. The detailed mechanism, however, is still not clear. Using live cell imaging techniques, we showed here that Bax underwent four distinct stages of dynamic redistribution during UV-induced apoptosis. At stage I, Bax was distributed diffusely in the cytosol. About an hour after UV treatment at stage II, Bax started to translocate to mitochondria and distributed uniformly at the mitochondrial outer membrane (MOM). Within a few minutes, at stage III, Bax and Bak began to form small complexes at the MOM. Later, at stage IV, these Bax and Bak complexes expanded to become large clusters. We found that the formation of Bax-Bak small complexes at stage III was responsible for permeabilizing the MOM to release cytochrome c and Smac. Using a FRET technique, we further showed that Bax binds to Bak within the complex formed at the MOM during stage III. Finally, using a quantitative fluorescence measurement, we determined that the Bax-Bak complex was about 0.25 microm wide and composed of more than 100 protein molecules. These findings suggest that the Bax-Bak structure responsible for releasing mitochondrial proteins during apoptosis is not channel-like.
Collapse
Affiliation(s)
- Liying Zhou
- Department of Biology, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | | |
Collapse
|
31
|
He L, Hou Z, Qi RZ. Calmodulin Binding and Cdk5 Phosphorylation of p35 Regulate Its Effect on Microtubules. J Biol Chem 2008; 283:13252-60. [DOI: 10.1074/jbc.m706937200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
32
|
A whole-cell assay for the high throughput screening of calmodulin antagonists. Anal Bioanal Chem 2008; 390:2073-9. [PMID: 18317736 DOI: 10.1007/s00216-008-1983-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 01/02/2008] [Accepted: 02/14/2008] [Indexed: 10/22/2022]
Abstract
Cell-based screening systems for pharmaceuticals are desired over molecular biosensing systems because of the information they provide on toxicity and bioavailability. However, the majority of sensing systems developed are molecular biosensing type screening systems and cannot be easily adapted to cell-based screening. In this study, we demonstrate that protein-based molecular sensing systems that employ a fluorescent protein as a signal transducer are amenable to cell-based sensing by expressing the protein molecular sensing system in the cell and employing these cells for screening of desired molecules. To achieve this, we expressed a molecular sensing system based on the fusion protein of calmodulin (CaM) and enhanced green fluorescent protein (EGFP) in bacterial cells, and utilized these cells for the screening of CaM antagonists. In the presence of Ca(2+), CaM undergoes a conformational change exposing a hydrophobic pocket that interacts with CaM-binding proteins, peptides, and drugs. This conformational change induced in CaM leads to a change in the microenvironment of EGFP, resulting in a change in its fluorescence intensity. The observed change in fluorescence intensity of EGFP can be correlated to the concentration of the analyte present in the sample. Dose-response curves for various tricyclic antidepressants were generated using cells containing CaM-EGFP fusion protein. Additionally, we demonstrate the versatility of our system for studying protein-protein interactions by using cells to study the binding of a peptide to CaM. The study showed that the CaM-EGFP fusion protein within the intact cells responds similarly to that of the isolated fusion protein, hence eliminating the need for any isolation and purification steps. We have demonstrated that this system can be used for the rapid screening of various CaM antagonists that are potential antipsychotic drugs.
Collapse
|
33
|
Batchelder EL, Thomas-Virnig CL, Hardin JD, White JG. Cytokinesis is not controlled by calmodulin or myosin light chain kinase in the Caenorhabditis elegans early embryo. FEBS Lett 2007; 581:4337-41. [PMID: 17716666 PMCID: PMC2144740 DOI: 10.1016/j.febslet.2007.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 07/27/2007] [Accepted: 08/03/2007] [Indexed: 12/16/2022]
Abstract
Furrow ingression in animal cell cytokinesis is controlled by phosphorylation of myosin II regulatory light chain (mRLC). In Caenorhabditis elegans embryos, Rho-dependent Kinase (RhoK) is involved in, but not absolutely required for, this phosphorylation. The calmodulin effector myosin light chain kinase (MLCK) can also phosphorylate mRLC and is widely regarded as a candidate for redundant function with RhoK. However, our results show that RNA mediated interference against C. elegans calmodulin and candidate MLCKs had no effect on cytokinesis in wild-type or RhoK mutant embryos, ruling out the calmodulin/MLCK pathway as the missing regulator of cytokinesis in the C. elegans early embryo.
Collapse
Affiliation(s)
- Ellen L Batchelder
- Laboratory of Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI 53706, USA
| | | | | | | |
Collapse
|
34
|
Yuan J, Shi GX, Shao Y, Dai G, Wei JN, Chang DC, Li CJ. Calmodulin bound to stress fibers but not microtubules involves regulation of cell morphology and motility. Int J Biochem Cell Biol 2007; 40:284-93. [PMID: 17884685 DOI: 10.1016/j.biocel.2007.08.004] [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] [Received: 04/10/2007] [Revised: 07/28/2007] [Accepted: 08/05/2007] [Indexed: 01/11/2023]
Abstract
Calmodulin (CaM) is a major cytoplasmic calcium receptor that performs multiple functions including cell motility. To investigate the mechanism of the regulation of CaM on cell morphology and motility, first we checked the distribution of CaM in the living cells using GFP-CaM as an indicator. We found that GFP-CaM showed a fiber-like distribution pattern in the cytosol of living Potorous tridactylis kidney (PtK2) cells but not in living HeLa cells. The endogenous CaM in heavily permeabilized HeLa was also found to display a fiber-like distribution pattern. Further examination showed that the distribution pattern of GFP-CaM was same as that of stress fibers, but not microtubules. Co-immunoprecipitation also showed that CaM can interact with actin directly or indirectly. The microinjection of trp peptide, a specific inhibitor of CaM, attenuated the polymerization of stress fibers and induced the alteration of cell morphology. A wound-healing assay and a single cell tracking experiment showed that CaM in PtK2 cells could increase cell motility. The data we have got from living cells suggested that CaM affect cell morphology and motility through binding to stress fibers and regulate f-actin polymerization.
Collapse
Affiliation(s)
- Jun Yuan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, #1 Wen Yuan Road, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Shen WG, Peng WX, Shao Y, Xu JF, Dai G, Zhang Y, Pan FY, Li CJ. Localization and activity of calmodulin is involved in cell–cell adhesion of tumor cells and endothelial cells in response to hypoxic stress. Cell Biol Toxicol 2007; 23:323-35. [PMID: 17351827 DOI: 10.1007/s10565-006-0157-2] [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] [Received: 07/23/2006] [Accepted: 11/29/2006] [Indexed: 01/11/2023]
Abstract
Adhesion of tumor cells to endothelial cells is known to be involved in the hematogenous metastasis of cancer, which is regulated by hypoxia. Hypoxia is able to induce a significant increase in free intracellular Ca2+ levels in both tumor cells and endothelial cells. Here, we investigate the regulatory effects of calmodulin (CaM), an intracellular calcium mediator, on tumor cell-endothelial cell adhesion under hypoxic conditions. Hypoxia facilitates HeLa cell-ECV304 endothelial cell adhesion, and results in actin cytoskeleton rearrangement in both endothelial cells and tumor cells. Suppression of CaM activation by CaM inhibitor W-7 disrupts actin cytoskeleton organization and CaM distribution in the cell-cell contact region, and thus inhibits cell-cell adhesion. CaM inhibitor also downregulates hypoxia-induced HIF-1-dependent gene expression. These results suggest that the Ca2+ -CaM signaling pathway might be involved in tumor cell-endothelial cell adhesion, and that co-localization of CaM and actin at cell-cell contact regions might be essential for this process under hypoxic stress.
Collapse
Affiliation(s)
- W-G Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Nanjing Normal University, Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Je HS, Yang F, Zhou J, Lu B. Neurotrophin 3 induces structural and functional modification of synapses through distinct molecular mechanisms. ACTA ACUST UNITED AC 2007; 175:1029-42. [PMID: 17178914 PMCID: PMC2064711 DOI: 10.1083/jcb.200603061] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanisms by which neurotrophins elicit long-term structural and functional changes of synapses are not known. We report the mechanistic separation of functional and structural synaptic regulation by neurotrophin 3 (NT-3), using the neuromuscular synapse as a model. Inhibition of cAMP response element (CRE)-binding protein (CREB)-mediated transcription blocks the enhancement of transmitter release elicited by NT-3, without affecting the synaptic varicosity of the presynaptic terminals. Further analysis indicates that CREB is activated through Ca(2+)/calmodulin-dependent kinase IV (CaMKIV) pathway, rather than the mitogen-activated protein kinase (MAPK) or cAMP pathway. In contrast, inhibition of MAPK prevents the NT-3-induced structural, but not functional, changes. Genetic and imaging experiments indicate that the small GTPase Rap1, but not Ras, acts upstream of MAPK activation by NT-3. Thus, NT-3 initiates parallel structural and functional modifications of synapses through the Rap1-MAPK and CaMKIV-CREB pathways, respectively. These findings may have implications in the general mechanisms of long-term synaptic modulation by neurotrophins.
Collapse
Affiliation(s)
- Hyun-Soo Je
- Section on Neural Development and Plasticity, National Institute of Child Health and Human Development, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
37
|
Eppinga RD, Li Y, Lin JLC, Lin JJC. Tropomyosin and caldesmon regulate cytokinesis speed and membrane stability during cell division. Arch Biochem Biophys 2006; 456:161-74. [PMID: 16854366 DOI: 10.1016/j.abb.2006.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Revised: 06/14/2006] [Accepted: 06/15/2006] [Indexed: 01/11/2023]
Abstract
The contractile ring and the cell cortex generate force to divide the cell while maintaining symmetrical shape. This requires temporal and spatial regulation of the actin cytoskeleton at these areas. We force-expressed misregulated versions of actin-binding proteins, tropomyosin and caldesmon, into cells and analyzed their effects on cell division. Cells expressing proteins that increase actomyosin ATPase, such as human tropomyosin chimera (hTM5/3), significantly speed up division, whereas cells expressing proteins that inhibit actomyosin, such as caldesmon mutants defective in Ca(2+)/calmodulin binding (CaD39-AB) and in cdk1 phosphorylation sites (CaD39-6F), divide slowly. hTM5 and hTM5/3-expressing cells lift one daughter cell off the substrate and twist. Furthermore, CaD39-AB- and CaD39-6F-expressing cells are sensitive to hypotonic swelling and show severe blebbing during division, whereas hTM5/3-expressing cells are resistant to hypotonic swelling and produce membrane bulges. These results support a model where Ca(2+)/calmodulin and cdk1 dynamically control caldesmon inhibition of tropomyosin-activated actomyosin to regulate division speed and to suppress membrane blebs.
Collapse
Affiliation(s)
- Robbin D Eppinga
- Department of Biological Sciences, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
38
|
Chen J, Dai G, Wang YQ, Wang S, Pan FY, Xue B, Zhao DH, Li CJ. Polo-like kinase 1 regulates mitotic arrest after UV irradiation through dephosphorylation of p53 and inducing p53 degradation. FEBS Lett 2006; 580:3624-30. [PMID: 16753148 DOI: 10.1016/j.febslet.2006.05.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 05/17/2006] [Accepted: 05/18/2006] [Indexed: 01/10/2023]
Abstract
Ultraviolet (UV) irradiation can result in cell cycle arrest. The reactivation of Polo-like kinase 1 (Plk1) is necessary for cell cycle reentry. But the mechanism of how Plk1 regulates p53 in UV-induced mitotic arrest cells remained elusive. Here we find that UV treatment leads HEK293 cells to inverse changes of Plk1 and p53. Over-expression of Plk1 rescue UV-induced mitotic arrest cells by inhibiting p53 activation. Plk1 could also inhibit p53 phosphorylation at Ser15, thus facilitates its nuclear export and degradation. Further examination shows that Plk1, p53 and Cdc25C can form a large complex. Plk1 could bind to the sequence-specific DNA-binding domain of p53 and active Cdc25C by hyperphosphorylation. These results hypothesize that Plk1 and Cdc25C participate in recovery the mitotic arrest through binding to the different domain of p53. Cdc25C may first be actived by Plk1, and then its phosphatase activity makes p53 dephosphorylated at Ser15.
Collapse
Affiliation(s)
- Jie Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, Life Sciences College, Nanjing Normal University, Nanjing 210097, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Tsang WY, Spektor A, Luciano DJ, Indjeian VB, Chen Z, Salisbury JL, Sánchez I, Dynlacht BD. CP110 cooperates with two calcium-binding proteins to regulate cytokinesis and genome stability. Mol Biol Cell 2006; 17:3423-34. [PMID: 16760425 PMCID: PMC1525247 DOI: 10.1091/mbc.e06-04-0371] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The centrosome is an integral component of the eukaryotic cell cycle machinery, yet very few centrosomal proteins have been fully characterized to date. We have undertaken a series of biochemical and RNA interference (RNAi) studies to elucidate a role for CP110 in the centrosome cycle. Using a combination of yeast two-hybrid screens and biochemical analyses, we report that CP110 interacts with two different Ca2+-binding proteins, calmodulin (CaM) and centrin, in vivo. In vitro binding experiments reveal a direct, robust interaction between CP110 and CaM and the existence of multiple high-affinity CaM-binding domains in CP110. Native CP110 exists in large (approximately 300 kDa to 3 MDa) complexes that contain both centrin and CaM. We investigated a role for CP110 in CaM-mediated events using RNAi and show that its depletion leads to a failure at a late stage of cytokinesis and the formation of binucleate cells, mirroring the defects resulting from ablation of either CaM or centrin function. Importantly, expression of a CP110 mutant unable to bind CaM also promotes cytokinesis failure and binucleate cell formation. Taken together, our data demonstrate a functional role for CaM binding to CP110 and suggest that CP110 cooperates with CaM and centrin to regulate progression through cytokinesis.
Collapse
Affiliation(s)
- William Y. Tsang
- *Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Alexander Spektor
- *Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Daniel J. Luciano
- *Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Vahan B. Indjeian
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | | | | | - Irma Sánchez
- *Department of Pathology, New York University School of Medicine, New York, NY 10016
| | - Brian David Dynlacht
- *Department of Pathology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
40
|
Sutherland CM, Moretti PAB, Hewitt NM, Bagley CJ, Vadas MA, Pitson SM. The calmodulin-binding site of sphingosine kinase and its role in agonist-dependent translocation of sphingosine kinase 1 to the plasma membrane. J Biol Chem 2006; 281:11693-701. [PMID: 16522638 DOI: 10.1074/jbc.m601042200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Sphingosine kinases catalyze the formation of sphingosine 1-phosphate, a bioactive lipid involved in many aspects of cellular regulation, including the fundamental biological processes of cell growth and survival. A diverse range of cell agonists induce activation of human sphingosine kinase 1 (hSK1) and, commonly, its translocation to the plasma membrane. Although the activation of hSK1 in response to at least some agonists occurs directly via its phosphorylation at Ser225 by ERK1/2, many aspects governing the regulation of this phosphorylation and subsequent translocation remain unknown. Here, in an attempt to understand some of these processes, we have examined the known interaction of hSK1 with calmodulin (CaM). By using a combination of limited proteolysis, peptide interaction analysis, and site-directed mutagenesis, we have identified that the CaM-binding site of hSK1 resides in the region spanned by residues 191-206. Specifically, Phe197 and Leu198 are critically involved in the interaction because a version of hSK1 incorporating mutations of both Phe197 --> Ala and Leu198 --> Gln failed to bind CaM. We have also shown for the first time that human sphingosine kinase 2 (hSK2) binds CaM, and does so via a CaM binding region that is conserved with hSK1 because comparable mutations in hSK2 also ablate CaM binding to this protein. By using the CaM-binding-deficient version of hSK1, we have begun to elucidate the role of CaM in hSK1 regulation by demonstrating that disruption of the CaM-binding site ablates agonist-induced translocation of hSK1 from the cytoplasm to the plasma membrane, while having no effect on hSK1 phosphorylation and catalytic activation.
Collapse
Affiliation(s)
- Catherine M Sutherland
- Hanson Institute, Division of Human Immunology, Institute of Medical and Veterinary Science, Frome Road, University of Adelaide, Adelaide, South Australia 5000, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Miyara F, Han Z, Gao S, Vassena R, Latham KE. Non-equivalence of embryonic and somatic cell nuclei affecting spindle composition in clones. Dev Biol 2006; 289:206-17. [PMID: 16310175 DOI: 10.1016/j.ydbio.2005.10.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2005] [Revised: 10/12/2005] [Accepted: 10/17/2005] [Indexed: 11/20/2022]
Abstract
Cloning by nuclear transfer remains inefficient but is more efficient when nuclei from embryonic cells or embryonic stem cells (ECNT) are employed as compared with somatic cells (SCNT). The factors determining efficiency have not been elucidated. We find that somatic and embryonic nuclei differ in their ability to organize meiotic and mitotic spindles of normal molecular composition. Calmodulin, a component of meiotic and mitotic spindle chromosome complexes (SCCs), displays sharply reduced association with the SCC forming after SCNT but not ECNT. This defect persists in mitotic spindles at least through the second mitosis, despite abundant calmodulin expression in the cell, and correlates with slow chromosome congression. We propose that somatic cell nuclei lack factors needed to direct normal SCC formation in oocytes and early embryos. These results reveal a striking control of SCC formation by the transplanted nucleus and provide the first identified molecular correlate of donor stage-dependent restriction in nuclear potency.
Collapse
Affiliation(s)
- Faical Miyara
- The Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
42
|
Yu YY, Dai G, Pan FY, Chen J, Li CJ. Calmodulin regulates the post-anaphase reposition of centrioles during cytokinesis. Cell Res 2005; 15:548-52. [PMID: 16045818 DOI: 10.1038/sj.cr.7290324] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A transient postanaphase repositioning of the centriole is found to control the completion of cytokinesis. Using a green fluorescent protein-calmodulin fusion protein as a living cell probe, we have previously found that calmodulin is associated with the initiation and progression of cytokinesis. In this study, we further studied the effect of calmodulin on the repositioning of the centriole and subsequent cell cycle progression. When activity of calmodulin is inhibited, the regression of the centriole from the intercellular bridge to the cell center is blocked, and thus the completion of cell division is repressed and two daughter cells are linked by longer cell bridge in perturbed cells. W7 treatment during cytokinesis also results in unfinished cytokinesis and stopped G1 phase. These results suggest that calmodulin activity is required for centriole repositioning and can affect the completion of cytokinesis and cell cycle progression.
Collapse
Affiliation(s)
- Yue Yue Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University. Nanjing 210097, China
| | | | | | | | | |
Collapse
|
43
|
Fabbro M, Zhou BB, Takahashi M, Sarcevic B, Lal P, Graham ME, Gabrielli BG, Robinson PJ, Nigg EA, Ono Y, Khanna KK. Cdk1/Erk2- and Plk1-dependent phosphorylation of a centrosome protein, Cep55, is required for its recruitment to midbody and cytokinesis. Dev Cell 2005; 9:477-88. [PMID: 16198290 DOI: 10.1016/j.devcel.2005.09.003] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Revised: 08/15/2005] [Accepted: 09/01/2005] [Indexed: 11/30/2022]
Abstract
Centrosomes in mammalian cells have recently been implicated in cytokinesis; however, their role in this process is poorly defined. Here, we describe a human coiled-coil protein, Cep55 (centrosome protein 55 kDa), that localizes to the mother centriole during interphase. Despite its association with gamma-TuRC anchoring proteins CG-NAP and Kendrin, Cep55 is not required for microtubule nucleation. Upon mitotic entry, centrosome dissociation of Cep55 is triggered by Erk2/Cdk1-dependent phosphorylation at S425 and S428. Furthermore, Cep55 locates to the midbody and plays a role in cytokinesis, as its depletion by siRNA results in failure of this process. S425/428 phosphorylation is required for interaction with Plk1, enabling phosphorylation of Cep55 at S436. Cells expressing phosphorylation-deficient mutant forms of Cep55 undergo cytokinesis failure. These results highlight the centrosome as a site to organize phosphorylation of Cep55, enabling it to relocate to the midbody to function in mitotic exit and cytokinesis.
Collapse
Affiliation(s)
- Megan Fabbro
- Queensland Institute of Medical Research, P.O. Royal Brisbane Hospital, Brisbane, Queensland 4029, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Yu YY, Chen Y, Dai G, Chen J, Sun XM, Wen CJ, Zhao DH, Chang DC, Li CJ. The association of calmodulin with central spindle regulates the initiation of cytokinesis in HeLa cells. Int J Biochem Cell Biol 2005; 36:1562-72. [PMID: 15147735 DOI: 10.1016/j.biocel.2003.12.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2003] [Revised: 12/02/2003] [Accepted: 12/18/2003] [Indexed: 01/12/2023]
Abstract
Calmodulin is a major cytoplasmic calcium receptor that performs multiple functions in the cell including cytokinesis. Central spindle appears between separating chromatin masses after metaphase-anaphase transition. The interaction of microtubules from central spindle with cell cortex regulates the cleavage furrow formation. In this paper, we use green fluorescence protein (GFP)-tagged calmodulin as a living cell probe to examine the detailed dynamic redistribution and co-localization of calmodulin with central spindle during cytokinesis and the function of this distribution pattern in a tripolar HeLa cell model. We found that calmodulin is associated with spindle microtubules during mitosis and begins to aggregate with central spindle after anaphase initiation. The absence of either central spindle or central spindle-distributed calmodulin is correlated with the defect in the formation of cleavage furrow, where contractile ring-distributed CaM is also extinct. Further analysis found that both the assembly of central spindle and the formation of cleavage furrow are affected by the W7 treatment. The microtubule density of central spindle was decreased after the treatment. Only less than 10% of the synchronized cells enter cytokinesis when treated with 25 microM W7, and the completion time of furrow regression is also delayed from 10 min to at least 40 min. It is suggested that calmodulin plays a significant role in cytokinesis including furrow formation and regression, The understanding of the interaction between calmodulin and microtubules may give us insight into the mechanism through which calmodulin regulates cytokinesis.
Collapse
Affiliation(s)
- Yue-Yue Yu
- Jiangsu Key Laboratory for Molecular & Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing 210097, Jiangsu Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Psatha M, Koffer A, Erent M, Moss SE, Bolsover S. Calmodulin spatial dynamics in RBL-2H3 mast cells. Cell Calcium 2004; 36:51-9. [PMID: 15126056 DOI: 10.1016/j.ceca.2003.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2003] [Revised: 11/15/2003] [Accepted: 11/25/2003] [Indexed: 10/26/2022]
Abstract
A line of rat basophilic leukaemia (RBL) cells, a model of mast cells, stably expressing EGFP-tagged calmodulin secreted normally in response to standard agonists. As reported for other cell types, calmodulin was concentrated in the mitotic spindle poles of dividing cells. In unstimulated interphase cells calmodulin was concentrated in the cell cortex and at a single central location. Disruption of cortical actin eliminated the concentration of calmodulin at the cortex while the central calmodulin concentration was associated with an enrichment of tubulin and is likely to represent the centrosome. Following stimulation with either an agonist that crosslinks Fc receptors or co-application of phorbol ester and a calcium ionophore the interior of the cells lost calmodulin while cortical fluorescence became more pronounced but also less uniform. After stimulation discrete bright puncta of calmodulin-EGFP (CaM-EGFP) appeared in the cell interior. Puncta colocalised with moving lysotracker-labelled granules, suggesting that calmodulin may play a role in organising their transport. Our results show that in interphase RBL cells a large fraction of the calmodulin pool is associated with targets in the actin cytoskeleton and demonstrate the utility of this model system for studying calmodulin biology.
Collapse
Affiliation(s)
- Maria Psatha
- Department of Physiology, University College London, London, UK
| | | | | | | | | |
Collapse
|
46
|
Thorogate R, Török K. Ca2+-dependent and -independent mechanisms of calmodulin nuclear translocation. J Cell Sci 2004; 117:5923-36. [PMID: 15522886 DOI: 10.1242/jcs.01510] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Translocation from the cytosol to the nucleus is a major response by calmodulin (CaM) to stimulation of cells by Ca2+. However, the mechanisms involved in this process are still controversial and both passive and facilitated diffusion have been put forward. We tested nuclear translocation mechanisms in electroporated HeLa cells, rat cortical neurons and glial cells using novel calmodulin and inhibitor peptide probes and confocal microscopy. Passive diffusion of calmodulin across the nuclear membrane was measured in conditions in which facilitated transport was blocked and was compared to that of a similarly sized fluorescein-labeled dextran. Wheat germ agglutinin, which blocks facilitated transport but not passive diffusion, inhibited the nuclear entry of both wild-type and Ca2+-binding-deficient mutant calmodulin both in low and elevated [Ca2+]. Ca2+-dependent nuclear translocation was prevented by a membrane-permeant CaM inhibitor, the mTrp peptide, which indicated that it was specific to Ca2+/CaM. Diffusion of free CaM and Ca2+/CaM was considerably slower than the observed nuclear translocation by facilitated transport. Our data show that the majority of CaM nuclear entry occurred by facilitated mechanisms in all cell types examined, in part by a Ca2+-independent and in part by a Ca2+-dependent translocation mechanism.
Collapse
Affiliation(s)
- Richard Thorogate
- Department of Basic Medical Sciences, St Georges Hospital Medical School, Cranmer Terrace, London, SW17 0RE, UK
| | | |
Collapse
|
47
|
Hoffman RM. In vivo imaging with fluorescent proteins: the new cell biology. Acta Histochem 2004; 106:77-87. [PMID: 15147628 DOI: 10.1016/j.acthis.2004.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2004] [Accepted: 02/08/2004] [Indexed: 10/26/2022]
Abstract
We propose a new cell biology where the behavior of cells can be visualized in the living animal. An example of the new cell biology is dual-color fluorescence imaging using red fluorescent protein (RFP)-expressing tumors transplanted in green fluorescent protein (GFP)-expressing transgenic mice. These models show with great clarity the details of tumor-stroma interactions and especially tumor-induced angiogenesis, tumor-infiltrating lymphocytes, stromal fibroblasts and macrophages. Another example is the color coding of cells with RFP or GFP such that both cell types can be simultaneously visualized in vivo. Stem cells can also be visualized and tracked in vivo. Mice in which the regulatory elements of the stem-cell marker nestin drive GFP enable nascent vasculature to be visualized interacting with transplanted RFP-expressing cancer cells. Nestin-driven GFP expression can also be used to visualize hair follicle stem cells. Dual-color cells expressing GFP in the nucleus and RFP in the cytoplasm enable real-time visualization of nuclear-cytoplasm dynamics including cell cycle events and apoptosis. Multiple-color labeling of cells will enable multiple events to be simultaneously visualized in vivo including gene expression, ion fluxes, protein and organelle trafficking, chromosome dynamics and numerous other processes currently still only studied in vitro.
Collapse
|
48
|
Nagasaki A, Uyeda TQP. DWWA, a novel protein containing two WW domains and an IQ motif, is required for scission of the residual cytoplasmic bridge during cytokinesis in Dictyostelium. Mol Biol Cell 2003; 15:435-46. [PMID: 14595117 PMCID: PMC329203 DOI: 10.1091/mbc.e03-05-0329] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
We have identified a novel gene, dwwA, which is required for cytokinesis of Dictyostelium cells on solid surfaces. Its product, Dd WW domain containing protein A (DWWA), contains several motifs, including two WW domains, an IQ motif, a C2 domain, and a proline-rich region. On substrates, cells lacking dwwA were multinucleated and larger and flatter than wild-type cells due to their frequent inability to sever the cytoplasmic bridge connecting daughter cells after mitosis. When cultured in suspension, however, dwwA-null cells seemed to carry out cytokinesis normally via a process not driven by the shearing force arising from agitation of the culture. GFP-DWWA localized to the cell cortex and nucleus; analysis of the distributions of various truncation mutants revealed that the N-terminal half of the protein, which contains the C2 domain, is required for the cortical localization of DWWA. The IQ motif of DWWA binds calmodulin in vitro. Given that the scission process is also defective in calmodulin knockdown cells cultured on substrates (Liu et al., 1992), we propose that DWWA's multiple binding domains enable it to function as an adaptor protein, facilitating the scission process through the regulation of Ca(2+)/calmodulin-mediated remodeling of the actin cytoskeleton and/or modulation of membrane dynamics.
Collapse
Affiliation(s)
- Akira Nagasaki
- Gene Function Research Center, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8562, Japan.
| | | |
Collapse
|
49
|
Popescu A, Miron S, Blouquit Y, Duchambon P, Christova P, Craescu CT. Xeroderma pigmentosum group C protein possesses a high affinity binding site to human centrin 2 and calmodulin. J Biol Chem 2003; 278:40252-61. [PMID: 12890685 DOI: 10.1074/jbc.m302546200] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human centrin 2 (HsCen2), a member of the EF-hand superfamily of Ca2+-binding proteins, is commonly associated with centrosome-related structures. The protein is organized in two domains, each containing two EF-hand motifs, but only the C-terminal half exhibits Ca2+ sensor properties. A significant fraction of HsCen2 is localized in the nucleus, where it was recently found associated with the xeroderma pigmentosum group C protein (XPC), a component of the nuclear excision repair pathway. Analysis of the XPC sequence (940 residues), using a calmodulin target recognition software, enabled us to predict two putative binding sites. The binding properties of the two corresponding peptides were investigated by isothermal titration calorimetry. Only one of the peptides (P1-XPC) interacts strongly (Ka = 2.2 x 10(8) m-1, stoichiometry 1:1) with HsCen2 in a Ca2+-dependent manner. This peptide also binds, with a similar affinity (Ka = 1.1 x 10(8) m-1) to a C-terminal construct of HsCen2, indicating that the interaction with the integral protein is mainly the result of the contribution of the C-terminal half. The second peptide (P2-XPC) failed to show any detectable binding either to HsCen2 or to its C-terminal lobe. The two peptides interact with different affinities and mechanisms with calmodulin. Circular dichroism and nuclear magnetic resonance were used to structurally characterize the complex formed by the C-terminal domain of HsCen2 with P1-XPC.
Collapse
Affiliation(s)
- Aurel Popescu
- INSERM U350 and Institut Curie-Recherche, Centre Universitaire, Batiments 110-112, 91405 Orsay, France
| | | | | | | | | | | |
Collapse
|
50
|
Okano H, Ohya Y. Binding of calmodulin to Nuf1p is required for karyogamy in Saccharomyces cerevisiae. Mol Genet Genomics 2003; 269:649-57. [PMID: 12836012 DOI: 10.1007/s00438-003-0853-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2003] [Accepted: 04/13/2003] [Indexed: 10/26/2022]
Abstract
The role of calmodulin (CaM) during mating in Saccharomyces cerevisiae was examined by using a set of Phe-to-Ala substitutions. We identified ten CaM mutants that exhibited significantly reduced mating efficiencies when crossed to a strain of the opposite mating type harboring the same CaM mutation. Most of the mating-defective CaM mutants were bilateral, i.e., they also exhibited mating defects, albeit minor ones, when crossed to the wild type. When strains carrying different bilateral CaM mutations were mated, the mating efficiencies recovered dramatically. We termed this phenomenon "intragenic mating complementation", and classified the mating-defective CaM mutations into two intragenic mating complementation groups. Two mutant alleles belonging to different groups showed minor defects in cell adhesion and cell fusion, but exhibited severe defects in karyogamy. CaM is known to bind to the essential spindle pole body component Nuf1p. This binding appears to be important for karyogamy because the nuf1(C911R) mutation, which impairs CaM-Nuf1p binding, resulted in a severe defect in karyogamy. Indeed, the two mating-defective CaM mutations were found to compromise formation of the CaM/Nuf1p complex, and the mating defects of these two CaM mutants were suppressible by a dominant, CaM-independent, mutation in NUF1. Taken together, these results suggest that loss of CaM binding to Nuf1p causes a defect in karyogamy, thereby inhibiting productive mating.
Collapse
Affiliation(s)
- H Okano
- Bio-Mimetic Control Research Center, The Institute of Physical and Chemical Research RIKEN, Anagahora, 463-0003 Nagoya, Aichi, Japan
| | | |
Collapse
|