251
|
Castellani CM, Torres-Ocampo AP, Breffke J, White AB, Chambers JJ, Stratton MM, Maresca TJ. Live-cell FLIM-FRET using a commercially available system. Methods Cell Biol 2020; 158:63-89. [PMID: 32423651 PMCID: PMC8006575 DOI: 10.1016/bs.mcb.2020.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Förster resonance energy transfer (FRET)-based sensors have been powerful tools in cell biologists' toolkit for decades. Informed by fundamental understanding of fluorescent proteins, protein-protein interactions, and the structural biology of reporter components, researchers have been able to employ creative design approaches to build sensors that are uniquely capable of probing a wide range of phenomena in living cells including visualization of localized calcium signaling, sub-cellular activity gradients, and tension generation to name but a few. While FRET sensors have significantly impacted many fields, one must also be cognizant of the limitations to conventional, intensity-based FRET measurements stemming from variation in probe concentration, sensitivity to photobleaching, and bleed-through between the FRET fluorophores. Fluorescence lifetime imaging microscopy (FLIM) largely overcomes the limitations of intensity-based FRET measurements. In general terms, FLIM measures the time, which for the reporters described in this chapter is nanoseconds (ns), between photon absorption and emission by a fluorophore. When FLIM is applied to FRET sensors (FLIM-FRET), measurement of the donor fluorophore lifetime provides valuable information such as FRET efficiency and the percentage of reporters engaged in FRET. This chapter introduces fundamental principles of FLIM-FRET toward informing the practical application of the technique and, using two established FRET reporters as proofs of concept, outlines how to use a commercially available FLIM system.
Collapse
Affiliation(s)
| | - Ana P. Torres-Ocampo
- Department of Biochemistry & Molecular Biology, University of Massachusetts, Amherst, MA,Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA
| | | | | | - James J. Chambers
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA
| | - Margaret M. Stratton
- Department of Biochemistry & Molecular Biology, University of Massachusetts, Amherst, MA,Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA
| | - Thomas J. Maresca
- Biology Department, University of Massachusetts, Amherst, MA,Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA
| |
Collapse
|
252
|
Osthole induces cell cycle arrest and apoptosis in head and neck squamous cell carcinoma by suppressing the PI3K/AKT signaling pathway. Chem Biol Interact 2019; 316:108934. [PMID: 31870840 DOI: 10.1016/j.cbi.2019.108934] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/14/2019] [Accepted: 12/19/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is one of the most common lethal tumors with a high recurrence rate and low survival rate. Therefore, an urgent need exists for novel and effective treatment strategies for HNSCC patients. METHODS Osthole, a natural ingredient extracted from Cnidium monnieri (L.) 'Cusson', has multiple pharmacological effects including antineoplastic activity. Regrettably, the antineoplastic effect of osthole in HNSCC cells remains undefined. We utilize in vitro assays to assess the anti-proliferative effects of osthole in HNSCC cells and tumorigenesis assays using FaDu cells in murine HNSCC models to assess in vivo function. Moreover, the possible molecular mechanisms of Osthole on HNSCC cells was also investigated. RESULTS Our findings show that the anti-proliferation effect of osthole might function through induction of cell cycle arrest (G2/M phase) and apoptosis in HNSCC. Osthole could also down-regulating the protein level of cell cycle and apoptosis related proteins, such as Bcl-2, PARP1, Survivin, CyclinB1 and Cdc2, while up-regulating expression of Cleaved Caspase3/9, Cleaved PARP1 and Bax. Similarly, osthole suppressed the in vivo growth of FaDu cells in a subcutaneous tumor model. In terms of mechanism, our data show that osthole can suppress the PI3K/AKT pathway. CONCLUSIONS In the current study, our in vitro and in vivo assay showed the suppressive effect of Osthole on HNSCC cells through induce cell cycle arrest (G2/M phase) and apoptosis. Moreover, the action mechanisms of Osthole on proliferation related signaling pathways was disclosed. Our present study suggests that osthole might be used as an effective therapeutic agent for patients with HNSCC.
Collapse
|
253
|
Hunter FW, Barker HR, Lipert B, Rothé F, Gebhart G, Piccart-Gebhart MJ, Sotiriou C, Jamieson SMF. Mechanisms of resistance to trastuzumab emtansine (T-DM1) in HER2-positive breast cancer. Br J Cancer 2019; 122:603-612. [PMID: 31839676 PMCID: PMC7054312 DOI: 10.1038/s41416-019-0635-y] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
The HER2-targeted antibody-drug conjugate trastuzumab emtansine (T-DM1) is approved for the treatment of metastatic, HER2-positive breast cancer after prior trastuzumab and taxane therapy, and has also demonstrated efficacy in the adjuvant setting in incomplete responders to neoadjuvant therapy. Despite its objective activity, intrinsic and acquired resistance to T-DM1 remains a major clinical challenge. T-DM1 mediates its activity in a number of ways, encompassing HER2 signalling blockade, Fc-mediated immune response and payload-mediated microtubule poisoning. Resistance mechanisms relating to each of these features have been demonstrated, and we outline the findings of these studies in this review. In our overview of the substantial literature on T-DM1 activity and resistance, we conclude that the T-DM1 resistance mechanisms most strongly supported by the experimental data relate to dysfunctional intracellular metabolism of the construct and subversion of DM1-mediated cell killing. Loss of dependence on signalling initiated by HER2-HER2 homodimers is not substantiated as a resistance mechanism by clinical or experimental studies, and the impact of EGFR expression and tumour immunological status requires further investigation. These findings are instructive with respect to strategies that might overcome T-DM1 resistance, including the use of second-generation anti-HER2 antibody-drug conjugates that deploy alternative linker-payload chemistries.
Collapse
Affiliation(s)
- Francis W Hunter
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand. .,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| | - Hilary R Barker
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Barbara Lipert
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Françoise Rothé
- Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Géraldine Gebhart
- Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | | | - Christos Sotiriou
- Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.,Department of Pharmacology and Clinical Pharmacology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
254
|
Wu H, Chen L, Zhu F, Han X, Sun L, Chen K. The Cytotoxicity Effect of Resveratrol: Cell Cycle Arrest and Induced Apoptosis of Breast Cancer 4T1 Cells. Toxins (Basel) 2019; 11:toxins11120731. [PMID: 31847250 PMCID: PMC6950385 DOI: 10.3390/toxins11120731] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Resveratrol, a natural polyterpenoid, can scavenge reactive oxygen species in vivo to carry out the functions of antioxidation and antiaging. Resveratrol’s anti-cancer capability has attracted widespread attention, but its molecular mechanism has not been systematically explained. In this study, by comparing the activity of normal cell lines and cancer cell lines after treating with resveratrol, it was found that resveratrol has more significant cytotoxicity in cancer cell lines. Resveratrol could play a toxic role through inducing apoptosis of the cancer cell in a time- and concentration-dependent manner. A total of 330 significantly differential genes were identified through large-scale transcriptome sequencing, among which 103 genes were upregulated and 227 genes were downregulated. Transcriptome and qRT-PCR data proved that a large number of genes related to cell cycle were differentially expressed after the treatment of resveratrol. The changes of cell cycle phases at different time points after treating with resveratrol were further detected, and it was found that the cells were arrested in the S phase because of the percentage of cells in S phase increased and cells in G1/G0 phase decreased. In conclusion, resveratrol can inhibit the proliferation of 4T1 cancer cells by inhibiting cell cycle and inducing apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Keping Chen
- Correspondence: ; Tel./Fax: +86-511-88791923
| |
Collapse
|
255
|
Marchetti G, Tavosanis G. Modulators of hormonal response regulate temporal fate specification in the Drosophila brain. PLoS Genet 2019; 15:e1008491. [PMID: 31809495 PMCID: PMC6919624 DOI: 10.1371/journal.pgen.1008491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 12/18/2019] [Accepted: 10/24/2019] [Indexed: 12/03/2022] Open
Abstract
Neuronal diversity is at the core of the complex processing operated by the nervous system supporting fundamental functions such as sensory perception, motor control or memory formation. A small number of progenitors guarantee the production of this neuronal diversity, with each progenitor giving origin to different neuronal types over time. How a progenitor sequentially produces neurons of different fates and the impact of extrinsic signals conveying information about developmental progress or environmental conditions on this process represent key, but elusive questions. Each of the four progenitors of the Drosophila mushroom body (MB) sequentially gives rise to the MB neuron subtypes. The temporal fate determination pattern of MB neurons can be influenced by extrinsic cues, conveyed by the steroid hormone ecdysone. Here, we show that the activation of Transforming Growth Factor-β (TGF-β) signalling via glial-derived Myoglianin regulates the fate transition between the early-born α’β’ and the pioneer αβ MB neurons by promoting the expression of the ecdysone receptor B1 isoform (EcR-B1). While TGF-β signalling is required in MB neuronal progenitors to promote the expression of EcR-B1, ecdysone signalling acts postmitotically to consolidate theα’β’ MB fate. Indeed, we propose that if these signalling cascades are impaired α’β’ neurons lose their fate and convert to pioneer αβ. Conversely, an intrinsic signal conducted by the zinc finger transcription factor Krüppel-homolog 1 (Kr-h1) antagonises TGF-β signalling and acts as negative regulator of the response mediated by ecdysone in promoting α’β’ MB neuron fate consolidation. Taken together, the consolidation of α’β’ MB neuron fate requires the response of progenitors to local signalling to enable postmitotic neurons to sense a systemic signal. Throughout the development of the central nervous system (CNS), a vast number of neuronal types are produced with striking precision. The unique identity of each neuronal cell type and the great cellular complexity in the CNS are established by intricate gene regulatory networks. Disruption of these identity programs leads to neurodevelopmental disorders and defects in cognition. Here, we report an important regulatory mechanism involved in consolidating neuronal fate. We show that during brain development local signalling, derived from interactions between glial cells and neuronal progenitors, is required to promote the expression of a hormone receptor in immature neurons. The perception of a systemic hormonal cue in those postmitotic neurons is fundamental for the consolidation of their neuronal fate. In this context, we additionally uncover an intrinsic regulatory mechanism that coordinates the hormone response to maintain the final neuronal fate.
Collapse
Affiliation(s)
- Giovanni Marchetti
- Dynamics of neuronal circuits, German Center for Neurodegenerative Diseases (DZNE), Germany
- * E-mail: (GM); (GT)
| | - Gaia Tavosanis
- Dynamics of neuronal circuits, German Center for Neurodegenerative Diseases (DZNE), Germany
- LIMES-Institute, University of Bonn, Germany
- * E-mail: (GM); (GT)
| |
Collapse
|
256
|
Sechi S, Frappaolo A, Karimpour-Ghahnavieh A, Gottardo M, Burla R, Di Francesco L, Szafer-Glusman E, Schininà E, Fuller MT, Saggio I, Riparbelli MG, Callaini G, Giansanti MG. Drosophila Doublefault protein coordinates multiple events during male meiosis by controlling mRNA translation. Development 2019; 146:dev.183053. [PMID: 31645358 DOI: 10.1242/dev.183053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/21/2019] [Indexed: 12/31/2022]
Abstract
During the extended prophase of Drosophila gametogenesis, spermatocytes undergo robust gene transcription and store many transcripts in the cytoplasm in a repressed state, until translational activation of select mRNAs in later steps of spermatogenesis. Here, we characterize the Drosophila Doublefault (Dbf) protein as a C2H2 zinc-finger protein, primarily expressed in testes, that is required for normal meiotic division and spermiogenesis. Loss of Dbf causes premature centriole disengagement and affects spindle structure, chromosome segregation and cytokinesis. We show that Dbf interacts with the RNA-binding protein Syncrip/hnRNPQ, a key regulator of localized translation in Drosophila We propose that the pleiotropic effects of dbf loss-of-function mutants are associated with the requirement of dbf function for translation of specific transcripts in spermatocytes. In agreement with this hypothesis, Dbf protein binds cyclin B mRNA and is essential for translation of cyclin B in mature spermatocytes.
Collapse
Affiliation(s)
- Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Angela Karimpour-Ghahnavieh
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Marco Gottardo
- Dipartimento di Scienze della Vita, Università di Siena, 53100 Siena, Italy
| | - Romina Burla
- Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Laura Di Francesco
- Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Edith Szafer-Glusman
- Departments of Developmental Biology and Genetics, Stanford University School of Medicine, Stanford, CA 94305-5329, USA
| | - Eugenia Schininà
- Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Margaret T Fuller
- Departments of Developmental Biology and Genetics, Stanford University School of Medicine, Stanford, CA 94305-5329, USA
| | - Isabella Saggio
- Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | | | - Giuliano Callaini
- Dipartimento di Biotecnologie Mediche, Università di Siena, 53100 Siena, Italy
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| |
Collapse
|
257
|
Vicario M, Cieri D, Vallese F, Catoni C, Barazzuol L, Berto P, Grinzato A, Barbieri L, Brini M, Calì T. A split-GFP tool reveals differences in the sub-mitochondrial distribution of wt and mutant alpha-synuclein. Cell Death Dis 2019; 10:857. [PMID: 31719530 PMCID: PMC6851186 DOI: 10.1038/s41419-019-2092-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/11/2019] [Accepted: 10/28/2019] [Indexed: 01/01/2023]
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disorder, is characterized by dopaminergic neuronal loss that initiates in the substantia nigra pars compacta and by the formation of intracellular inclusions mainly constituted by aberrant α-synuclein (α-syn) deposits known as Lewy bodies. Most cases of PD are sporadic, but about 10% are familial, among them those caused by mutations in SNCA gene have an autosomal dominant transmission. SNCA encodes α-syn, a small 140-amino acids protein that, under physiological conditions, is mainly localized at the presynaptic terminals. It is prevalently cytosolic, but its presence has been reported in the nucleus, in the mitochondria and, more recently, in the mitochondria-associated ER membranes (MAMs). Whether different cellular localizations may reflect specific α-syn activities is presently unclear and its action at mitochondrial level is still a matter of debate. Mounting evidence supports a role for α-syn in several mitochondria-derived activities, among which maintenance of mitochondrial morphology and modulation of complex I and ATP synthase activity. α-syn has been proposed to localize at the outer membrane (OMM), in the intermembrane space (IMS), at the inner membrane (IMM) and in the mitochondrial matrix, but a clear and comparative analysis of the sub-mitochondrial localization of WT and mutant α-syn is missing. Furthermore, the reasons for this spread sub-mitochondrial localization under physiological and pathological circumstances remain elusive. In this context, we decided to selectively monitor the sub-mitochondrial distribution of the WT and PD-related α-syn mutants A53T and A30P by taking advantage from a bimolecular fluorescence complementation (BiFC) approach. We also investigated whether cell stress could trigger α-syn translocation within the different mitochondrial sub-compartments and whether PD-related mutations could impinge on it. Interestingly, the artificial targeting of α-syn WT (but not of the mutants) to the mitochondrial matrix impacts on ATP production, suggesting a potential role within this compartment.
Collapse
Affiliation(s)
- Mattia Vicario
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Domenico Cieri
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Francesca Vallese
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Lucia Barazzuol
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paola Berto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Laura Barbieri
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Biology, University of Padova, Padova, Italy.
| | - Tito Calì
- Department of Biomedical Sciences, University of Padova, Padova, Italy. .,Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
| |
Collapse
|
258
|
lncRNA Expression Reveals the Potential Regulatory Roles in Hepatocyte Proliferation during Rat Liver Regeneration. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8597953. [PMID: 31828136 PMCID: PMC6885160 DOI: 10.1155/2019/8597953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022]
Abstract
Liver regeneration is a tissue growth process after loss or injury of liver tissue, which is a compensatory hyperplasia rather than true regeneration, mainly depending on hepatocyte proliferation. Currently, a large number of studies on hepatocyte proliferation have been conducted. However, studies on the regulation of long noncoding RNA (lncRNA) on hepatocyte proliferation are still limited. To identify specially expressed lncRNA during rat liver regeneration, high-throughput sequencing technology was performed, and a total of 2446 lncRNAs and 4091 mRNAs were identified as significantly differentially expressed. Gene ontology (GO) enrichment analysis was performed to analyze the role of differentially expressed mRNAs, and 695 mRNAs were identified to be related to cell proliferation. Then, an lncRNA-mRNA coexpression network based on the differentially expressed lncRNAs and proliferation-related genes was constructed to analyze the potential function of lncRNAs on hepatocyte proliferation, and ten lncRNAs, NONRATT003557.2, NONRATT005357.2, NONRATT003292.2, NONRATT001466.2, NONRATT003289.2, NONRATT001047.2, NONRATT005180.2, NONRATT004419.2, NONRATT005336.2, and NONRATT005335.2, were selected as key regulatory factors, which may play crucial roles in hepatocyte proliferation during rat liver regeneration. Finally, a protein-protein interaction (PPI) network was established to illuminate the interaction between proliferation-related genes, and ten hub genes (Aurkb, Cdk1, Cdc20, Bub1b, Mad2l1, Kif11, Prc1, Ccna2, Top2a, and Ccnb1) were screened with the MCC method in the PPI network, which may be important biomarkers involved in the hepatocyte proliferation during rat liver regeneration. These results may provide clues for a more comprehensive understanding of the molecular mechanism of hepatocyte proliferation during rat liver regeneration.
Collapse
|
259
|
Karnay A, Karisetty BC, Beaver M, Elefant F. Hippocampal stimulation promotes intracellular Tip60 dynamics with concomitant genome reorganization and synaptic gene activation. Mol Cell Neurosci 2019; 101:103412. [PMID: 31682915 DOI: 10.1016/j.mcn.2019.103412] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/07/2019] [Accepted: 09/17/2019] [Indexed: 11/17/2022] Open
Abstract
Genomic reorganizations mediating the engagement of target genes to transcription factories (TFs), characterized as specialized nuclear subcompartments enriched in hyperphosphorylated RNA polymerase II (RNAPII) and transcriptional regulators, act as an important layer of control in coordinating efficient gene transcription. However, their presence in hippocampal neurons and potential role in activity-dependent coregulation of genes within the brain remains unclear. Here, we investigate whether the well-characterized role for the histone acetyltransferase (HAT) Tip60 in mediating epigenetic control of inducible neuroplasticity genes involves TF associated chromatin reorganization in the hippocampus. We show that Tip60 shuttles into the nucleus following extracellular stimulation of rat hippocampal neurons with concomitant enhancement of Tip60 binding and activation of specific synaptic plasticity genes. Multicolor three-dimensional (3D) DNA fluorescent in situ hybridization (DNA-FISH) reveals that hippocampal stimulation mobilizes these same synaptic plasticity genes and Tip60 to RNAPII-rich TFs. Our data support a model by which external hippocampal stimulation promotes intracellular Tip60 HAT dynamics with concomitant TF associated genome reorganization to initiate Tip60mediated synaptic gene activation.
Collapse
Affiliation(s)
- Ashley Karnay
- Department of Biology, Drexel University, Philadelphia, PA, USA; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | - Mariah Beaver
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
260
|
Song W, Guo C, Chen J, Duan S, Hu Y, Zou Y, Chi H, Geng J, Zhou J. Silencing PSME3 induces colorectal cancer radiosensitivity by downregulating the expression of cyclin B1 and CKD1. Exp Biol Med (Maywood) 2019; 244:1409-1418. [PMID: 31630568 DOI: 10.1177/1535370219883408] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Resistance to radiotherapy remains a severe obstacle in the treatment of high-risk colorectal cancer patients. Recent studies have indicated that proteasome activator complex subunit 3 (PSME3) participates in the development and progression of various human malignancies and is proposed to play a role in tumor radioresistance. However, the impact of PSME3 on radioresistance of colorectal cancer has been largely unknown. In the present study, the enhanced expression of PSME3 was observed in colorectal cancer cells and tissue. Upregulation of PSME3 was significantly implicated in lymph node state, lymphovascular invasion, and Dukes' stage. Furthermore, high PSME3 expression was closely linked to poorer overall and progression-free survival in patients with colorectal cancer. The study further demonstrated that the proliferative, invasive and migratory potential of colorectal cancer cells was effectively inhibited in vitro after silencing PSME3. Our results verified that knockdown of PSME3 probably triggered cell cycle arrest at the G2/M phase by downregulation of cyclinB1 and CDK1, thereby enhancing the radiosensitivity of colorectal cancer cells. These data illustrated that PSME3 is a promising biomarker predictive of colorectal cancer prognosis and silencing of PSME3 may provide with a new approach for sensitizing the radiotherapy in colorectal cancer. Impact statement It is reported that colorectal cancer (CRC) is the third most common cancer worldwide and the fourth leading cause of cancer-related death. At present, the main treatment method of colorectal cancer is surgery, supplemented by radiotherapy and chemotherapy. Among them, radiotherapy plays an important role in the treatment of locally advanced colorectal cancer, surgery, and chemotherapy. Our study found that down-regulation of PSME3 may enhance the radiosensitivity of CRC cells by triggering cell cycle arrest, which suggests that silence PSME3 may provide a new method for improving the radiosensitivity of CRC. What’more, our research also demonstrated that PSME3 may promote proliferation, invasive and migratory potential of CRC cells, which implies that PSME3 might be a biomarker of CRC for early diagnosis and treatment.
Collapse
Affiliation(s)
- Wen Song
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Cuiping Guo
- Department of Emergency, Zhumadian Second Hospital of Traditional Chinese Medicine, Zhumadian 463000, China
| | - Jianxiong Chen
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shiyu Duan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yukun Hu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ying Zou
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second School of Clinical Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Honggang Chi
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second School of Clinical Medicine, Guangdong Medical University, Dongguan 523808, China
| | - Jian Geng
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jun Zhou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
261
|
Abstract
Chromosome segregation errors in human oocytes lead to aneuploid embryos that cause infertility and birth defects. Here we provide an overview of the chromosome-segregation process in the mammalian oocyte, highlighting mechanistic differences between oocytes and somatic cells that render oocytes so prone to segregation error. These differences include the extremely large size of the oocyte cytoplasm, the unique geometry of meiosis-I chromosomes, idiosyncratic function of the spindle assembly checkpoint, and dramatically altered oocyte cell-cycle control and spindle assembly, as compared to typical somatic cells. We summarise recent work suggesting that aging leads to a further deterioration in fidelity of chromosome segregation by impacting multiple components of the chromosome-segregation machinery. In addition, we compare and contrast recent results from mouse and human oocytes, which exhibit overlapping defects to differing extents. We conclude that the striking propensity of the oocyte to mis-segregate chromosomes reflects the unique challenges faced by the spindle in a highly unusual cellular environment.
Collapse
Affiliation(s)
- Aleksandar I Mihajlović
- Centre Recherche CHUM and Department OBGYN, Université de Montreal, Montreal, Quebec, Canada
| | - Greg FitzHarris
- Centre Recherche CHUM and Department OBGYN, Université de Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
262
|
Crncec A, Hochegger H. Triggering mitosis. FEBS Lett 2019; 593:2868-2888. [PMID: 31602636 DOI: 10.1002/1873-3468.13635] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/28/2022]
Abstract
Entry into mitosis is triggered by the activation of cyclin-dependent kinase 1 (Cdk1). This simple reaction rapidly and irreversibly sets the cell up for division. Even though the core step in triggering mitosis is so simple, the regulation of this cellular switch is highly complex, involving a large number of interconnected signalling cascades. We do have a detailed knowledge of most of the components of this network, but only a poor understanding of how they work together to create a precise and robust system that ensures that mitosis is triggered at the right time and in an orderly fashion. In this review, we will give an overview of the literature that describes the Cdk1 activation network and then address questions relating to the systems biology of this switch. How is the timing of the trigger controlled? How is mitosis insulated from interphase? What determines the sequence of events, following the initial trigger of Cdk1 activation? Which elements ensure robustness in the timing and execution of the switch? How has this system been adapted to the high levels of replication stress in cancer cells?
Collapse
Affiliation(s)
- Adrijana Crncec
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Helfrid Hochegger
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| |
Collapse
|
263
|
Wang Y, Sun J, Ni Q, Nie A, Gu Y, Wang S, Zhang W, Ning G, Wang W, Wang Q. Dual Effect of Raptor on Neonatal β-Cell Proliferation and Identity Maintenance. Diabetes 2019; 68:1950-1964. [PMID: 31345937 DOI: 10.2337/db19-0166] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/11/2019] [Indexed: 11/13/2022]
Abstract
Immature pancreatic β-cells are highly proliferative, and the expansion of β-cells during the early neonatal period largely determines functional β-cell mass; however, the mechanisms are poorly characterized. We generated Ngn3RapKO mice (ablation of Raptor, an essential component of mechanistic target of rapamycin [mTORC1] in Ngn3+ endocrine progenitor cells) and found that mTORC1 was dispensable for endocrine cell lineage formation but specifically regulated both proliferation and identity maintenance of neonatal β-cells. Ablation of Raptor in neonatal β-cells led to autonomous loss of cell identity, decelerated cell cycle progression, compromised proliferation, and caused neonatal diabetes as a result of inadequate establishment of functional β-cell mass at postnatal day 14. Completely different from mature β-cells, Raptor regulated G1/S and G2/M phase cell cycle transition, thus permitting a high proliferation rate in neonatal β-cells. Moreover, Ezh2 was identified as a critical downstream target of mTORC1 in neonatal β-cells, which was responsible for G2/M phase transition and proliferation. Our discovery of the dual effect of mTORC1 in immature β-cells has revealed a potential target for replenishing functional β-cell pools by promoting both expansion and functional maturation of newly formed immature β-cells.
Collapse
Affiliation(s)
- Yanqiu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajun Sun
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qicheng Ni
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aifang Nie
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyun Gu
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, China
| | - Guang Ning
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qidi Wang
- Shanghai Institute of Endocrine and Metabolic Diseases, Department of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors and E-Institute for Endocrinology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
264
|
Holder J, Poser E, Barr FA. Getting out of mitosis: spatial and temporal control of mitotic exit and cytokinesis by PP1 and PP2A. FEBS Lett 2019; 593:2908-2924. [PMID: 31494926 DOI: 10.1002/1873-3468.13595] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/31/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
Here, we will review the evidence showing that mitotic exit is initiated by regulated proteolysis and then driven by the PPP family of phosphoserine/threonine phosphatases. Rapid APC/CCDC20 and ubiquitin-dependent proteolysis of cyclin B and securin initiates sister chromatid separation, the first step of mitotic exit. Because proteolysis of Aurora and Polo family kinases dependent on APC/CCDH1 is relatively slow, this creates a new regulatory state, anaphase, different to G2 and M-phase. We will discuss how the CDK1-counteracting phosphatases PP1 and PP2A-B55, together with Aurora and Polo kinases, contribute to the temporal regulation and order of events in the different stages of mitotic exit from anaphase to cytokinesis. For PP2A-B55, these timing properties are created by the ENSA-dependent inhibitory pathway and differential recognition of phosphoserine and phosphothreonine. Finally, we will discuss how Aurora B and PP2A-B56 are needed for the spatial regulation of anaphase spindle formation and how APC/C-dependent destruction of PLK1 acts as a timer for abscission, the final event of cytokinesis.
Collapse
Affiliation(s)
- James Holder
- Department of Biochemistry, University of Oxford, UK
| | - Elena Poser
- Department of Biochemistry, University of Oxford, UK
| | | |
Collapse
|
265
|
The cell cycle in stem cell proliferation, pluripotency and differentiation. Nat Cell Biol 2019; 21:1060-1067. [PMID: 31481793 DOI: 10.1038/s41556-019-0384-4] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/24/2019] [Indexed: 12/30/2022]
Abstract
Cyclins, cyclin-dependent kinases and other components of the core cell cycle machinery drive cell division. Growing evidence indicates that this machinery operates in a distinct fashion in some mammalian stem cell types, such as pluripotent embryonic stem cells. In this Review, we discuss our current knowledge of how cell cycle proteins mechanistically link cell proliferation, pluripotency and cell fate specification. We focus on embryonic stem cells, induced pluripotent stem cells and embryonic neural stem/progenitor cells.
Collapse
|
266
|
Afonso O, Castellani CM, Cheeseman LP, Ferreira JG, Orr B, Ferreira LT, Chambers JJ, Morais-de-Sá E, Maresca TJ, Maiato H. Spatiotemporal control of mitotic exit during anaphase by an aurora B-Cdk1 crosstalk. eLife 2019; 8:e47646. [PMID: 31424385 PMCID: PMC6706241 DOI: 10.7554/elife.47646] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/10/2019] [Indexed: 11/13/2022] Open
Abstract
According to the prevailing 'clock' model, chromosome decondensation and nuclear envelope reformation when cells exit mitosis are byproducts of Cdk1 inactivation at the metaphase-anaphase transition, controlled by the spindle assembly checkpoint. However, mitotic exit was recently shown to be a function of chromosome separation during anaphase, assisted by a midzone Aurora B phosphorylation gradient - the 'ruler' model. Here we found that Cdk1 remains active during anaphase due to ongoing APC/CCdc20- and APC/CCdh1-mediated degradation of B-type Cyclins in Drosophila and human cells. Failure to degrade B-type Cyclins during anaphase prevented mitotic exit in a Cdk1-dependent manner. Cyclin B1-Cdk1 localized at the spindle midzone in an Aurora B-dependent manner, with incompletely separated chromosomes showing the highest Cdk1 activity. Slowing down anaphase chromosome motion delayed Cyclin B1 degradation and mitotic exit in an Aurora B-dependent manner. Thus, a crosstalk between molecular 'rulers' and 'clocks' licenses mitotic exit only after proper chromosome separation.
Collapse
Affiliation(s)
- Olga Afonso
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | | | - Liam P Cheeseman
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Jorge G Ferreira
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
- Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de MedicinaUniversidade do PortoPortoPortugal
| | - Bernardo Orr
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - Luisa T Ferreira
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
| | - James J Chambers
- Institute for Applied Life SciencesUniversity of MassachusettsAmherstUnited States
| | - Eurico Morais-de-Sá
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
- Epithelial Polarity & Cell Division Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
| | - Thomas J Maresca
- Biology DepartmentUniversity of MassachusettsAmherstUnited States
- Molecular and Cellular Biology Graduate ProgramUniversity of MassachusettsAmherstUnited States
| | - Helder Maiato
- Chromosome Instability & Dynamics Group, i3S - Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Instituto de Biologia Molecular e CelularUniversidade do PortoPortoPortugal
- Cell Division Group, Experimental Biology Unit, Department of Biomedicine, Faculdade de MedicinaUniversidade do PortoPortoPortugal
| |
Collapse
|
267
|
Jones MC, Zha J, Humphries MJ. Connections between the cell cycle, cell adhesion and the cytoskeleton. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180227. [PMID: 31431178 PMCID: PMC6627016 DOI: 10.1098/rstb.2018.0227] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Cell division, the purpose of which is to enable cell replication, and in particular to distribute complete, accurate copies of genetic material to daughter cells, is essential for the propagation of life. At a morphological level, division not only necessitates duplication of cellular structures, but it also relies on polar segregation of this material followed by physical scission of the parent cell. For these fundamental changes in cell shape and positioning to be achieved, mechanisms are required to link the cell cycle to the modulation of cytoarchitecture. Outside of mitosis, the three main cytoskeletal networks not only endow cells with a physical cytoplasmic skeleton, but they also provide a mechanism for spatio-temporal sensing via integrin-associated adhesion complexes and site-directed delivery of cargoes. During mitosis, some interphase functions are retained, but the architecture of the cytoskeleton changes dramatically, and there is a need to generate a mitotic spindle for chromosome segregation. An economical solution is to re-use existing cytoskeletal molecules: transcellular actin stress fibres remodel to create a rigid cortex and a cytokinetic furrow, while unipolar radial microtubules become the primary components of the bipolar spindle. This remodelling implies the existence of specific mechanisms that link the cell-cycle machinery to the control of adhesion and the cytoskeleton. In this article, we review the intimate three-way connection between microenvironmental sensing, adhesion signalling and cell proliferation, particularly in the contexts of normal growth control and aberrant tumour progression. As the morphological changes that occur during mitosis are ancient, the mechanisms linking the cell cycle to the cytoskeleton/adhesion signalling network are likely to be primordial in nature and we discuss recent advances that have elucidated elements of this link. A particular focus is the connection between CDK1 and cell adhesion. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
| | | | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
268
|
Vennin C, Mélénec P, Rouet R, Nobis M, Cazet AS, Murphy KJ, Herrmann D, Reed DA, Lucas MC, Warren SC, Elgundi Z, Pinese M, Kalna G, Roden D, Samuel M, Zaratzian A, Grey ST, Da Silva A, Leung W, Mathivanan S, Wang Y, Braithwaite AW, Christ D, Benda A, Parkin A, Phillips PA, Whitelock JM, Gill AJ, Sansom OJ, Croucher DR, Parker BL, Pajic M, Morton JP, Cox TR, Timpson P. CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan. Nat Commun 2019; 10:3637. [PMID: 31406163 PMCID: PMC6691013 DOI: 10.1038/s41467-019-10968-6] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
Heterogeneous subtypes of cancer-associated fibroblasts (CAFs) coexist within pancreatic cancer tissues and can both promote and restrain disease progression. Here, we interrogate how cancer cells harboring distinct alterations in p53 manipulate CAFs. We reveal the existence of a p53-driven hierarchy, where cancer cells with a gain-of-function (GOF) mutant p53 educate a dominant population of CAFs that establish a pro-metastatic environment for GOF and null p53 cancer cells alike. We also demonstrate that CAFs educated by null p53 cancer cells may be reprogrammed by either GOF mutant p53 cells or their CAFs. We identify perlecan as a key component of this pro-metastatic environment. Using intravital imaging, we observe that these dominant CAFs delay cancer cell response to chemotherapy. Lastly, we reveal that depleting perlecan in the stroma combined with chemotherapy prolongs mouse survival, supporting it as a potential target for anti-stromal therapies in pancreatic cancer.
Collapse
Affiliation(s)
- Claire Vennin
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
- Molecular Pathology department, the Netherlands Cancer Institute, Amsterdam, 1066CX, the Netherlands
| | - Pauline Mélénec
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Romain Rouet
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Max Nobis
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Aurélie S Cazet
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Kendelle J Murphy
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - David Herrmann
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Daniel A Reed
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Morghan C Lucas
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Sean C Warren
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Zehra Elgundi
- Graduate school of Biomedical Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Mark Pinese
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Gabriella Kalna
- Cancer Research UK Beatson Institute, Glasgow Scotland, G61 BD, UK
| | - Daniel Roden
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Monisha Samuel
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Anaiis Zaratzian
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
| | - Shane T Grey
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Andrew Da Silva
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
| | - Wilfred Leung
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Suresh Mathivanan
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, CA, 92121, USA
| | - Anthony W Braithwaite
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, 2006, Australia
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, 9054, New Zealand
- Maurice Wilkins Centre, University of Otago, Dunedin, 9054, New Zealand
| | - Daniel Christ
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Ales Benda
- Biomedical imaging facility, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW, Australia
| | - Ashleigh Parkin
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - John M Whitelock
- Graduate school of Biomedical Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Anthony J Gill
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards, Sydney, NSW, 2065, Australia
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow Scotland, G61 BD, UK
| | - David R Croucher
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | - Benjamin L Parker
- Schools of Life and Environmental Sciences, the Charles Perkin Centre, the University of Sydney, Sydney, NSW, 2006, Australia
| | - Marina Pajic
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia
| | | | - Thomas R Cox
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia.
| | - Paul Timpson
- The Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Sydney, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales Sydney, Sydney, NSW, 2010, Australia.
| |
Collapse
|
269
|
Interplay between Phosphatases and the Anaphase-Promoting Complex/Cyclosome in Mitosis. Cells 2019; 8:cells8080814. [PMID: 31382469 PMCID: PMC6721574 DOI: 10.3390/cells8080814] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/25/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Accurate division of cells into two daughters is a process that is vital to propagation of life. Protein phosphorylation and selective degradation have emerged as two important mechanisms safeguarding the delicate choreography of mitosis. Protein phosphatases catalyze dephosphorylation of thousands of sites on proteins, steering the cells through establishment of the mitotic phase and exit from it. A large E3 ubiquitin ligase, the anaphase-promoting complex/cyclosome (APC/C) becomes active during latter stages of mitosis through G1 and marks hundreds of proteins for destruction. Recent studies have revealed the complex interregulation between these two classes of enzymes. In this review, we highlight the direct and indirect mechanisms by which phosphatases and the APC/C mutually influence each other to ensure accurate spatiotemporal and orderly progression through mitosis, with a particular focus on recent insights and conceptual advances.
Collapse
|
270
|
Leung JC, Cassimeris L. Reorganization of paclitaxel-stabilized microtubule arrays at mitotic entry: roles of depolymerizing kinesins and severing proteins. Cancer Biol Ther 2019; 20:1337-1347. [PMID: 31345098 PMCID: PMC6783116 DOI: 10.1080/15384047.2019.1638678] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Paclitaxel is a widely used anti-cancer treatment that disrupts cell cycle progression by blocking cells in mitosis. The block at mitosis, with spindles assembled from short microtubules, is surprising given paclitaxel’s microtubule stabilizing activity and the need to depolymerize long interphase microtubules prior to spindle formation. Cells must antagonize paclitaxel’s microtubule stabilizing activity during a brief window of time at the transition from interphase to mitosis, allowing microtubule reorganization into a mitotic spindle, although the mechanism underlying microtubule depolymerization in the presence of paclitaxel has not been examined. Here we test the hypothesis that microtubule severing and/or depolymerizing proteins active at mitotic entry are necessary to clear the interphase array in paclitaxel-treated cells and allow subsequent formation of mitotic spindles formed of short microtubules. A549 and LLC-PK1 cells treated with 30nM paclitaxel approximately 4 h prior to mitotic entry successfully progress through the G2/M transition by clearing the interphase microtubule array from the cell interior outward to the cell periphery, a spatial pattern of reorganization that differs from that of cells possessing dynamic microtubules. Depletion of kinesin-8s, KIF18A and/or KIF18B obstructed interphase microtubule clearing at mitotic entry in paclitaxel-treated cells, with KIF18B making the larger contribution. Of the severing proteins, depletion of spastin, but not katanin, reduced microtubule loss as cells entered mitosis in the presence of paclitaxel. These results support a model in which KIF18A, KIF18B, and spastin promote interphase microtubule array disassembly at mitotic entry and can overcome paclitaxel-induced microtubule stability specifically at the G2/M transition.
Collapse
Affiliation(s)
- Jessica C Leung
- Department of Biological Sciences, 111 Research Dr. Lehigh University , Bethlehem , PA , USA
| | - Lynne Cassimeris
- Department of Biological Sciences, 111 Research Dr. Lehigh University , Bethlehem , PA , USA
| |
Collapse
|
271
|
Li Y, Guo L, Li H, Li J, Dong F, Yi Z, Ouyang Y, Hou Y, Wang Z, Sun Q, Lu S, Han Z. NEK5 regulates cell cycle progression during mouse oocyte maturation and preimplantation embryonic development. Mol Reprod Dev 2019; 86:1189-1198. [DOI: 10.1002/mrd.23234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Yuan‐Yuan Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, College of Animal Science and TechnologyGuangxi UniversityNanning China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Lei Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Hui Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Jian Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Feng Dong
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Zi‐Yun Yi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Ying‐Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Yi Hou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Zhen‐Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Qing‐Yuan Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| | - Sheng‐Sheng Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro‐Bioresources, Guangxi High Education Key Laboratory for Animal Reproduction and Biotechnology, College of Animal Science and TechnologyGuangxi UniversityNanning China
| | - Zhiming Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijing China
| |
Collapse
|
272
|
Mandati V, Del Maestro L, Dingli F, Lombard B, Loew D, Molinie N, Romero S, Bouvard D, Louvard D, Gautreau AM, Pasmant E, Lallemand D. Phosphorylation of Merlin by Aurora A kinase appears necessary for mitotic progression. J Biol Chem 2019; 294:12992-13005. [PMID: 31296571 DOI: 10.1074/jbc.ra118.006937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 06/25/2019] [Indexed: 01/01/2023] Open
Abstract
Although Merlin's function as a tumor suppressor and regulator of mitogenic signaling networks such as the Ras/rac, Akt, and Hippo pathways is well-documented, in mammals as well as in insects, its role during cell cycle progression remains unclear. In this study, using a combination of approaches, including FACS analysis, time-lapse imaging, immunofluorescence microscopy, and co-immunoprecipitation, we show that Ser-518 of Merlin is a substrate of the Aurora protein kinase A during mitosis and that its phosphorylation facilitates the phosphorylation of a newly discovered site, Thr-581. We found that the expression in HeLa cells of a Merlin variant that is phosphorylation-defective on both sites leads to a defect in centrosomes and mitotic spindles positioning during metaphase and delays the transition from metaphase to anaphase. We also show that the dual mitotic phosphorylation not only reduces Merlin binding to microtubules but also timely modulates ezrin interaction with the cytoskeleton. Finally, we identify several point mutants of Merlin associated with neurofibromatosis type 2 that display an aberrant phosphorylation profile along with defective α-tubulin-binding properties. Altogether, our findings of an Aurora A-mediated interaction of Merlin with α-tubulin and ezrin suggest a potential role for Merlin in cell cycle progression.
Collapse
Affiliation(s)
- Vinay Mandati
- CNRS, UMR144, Institute Curie, PSL Research University, F-75005 Paris, France
| | | | - Florent Dingli
- Laboratoire de Spectrométrie de Masse Protéomique, Institute Curie, PSL Research University, Paris, France
| | - Bérangère Lombard
- Laboratoire de Spectrométrie de Masse Protéomique, Institute Curie, PSL Research University, Paris, France
| | - Damarys Loew
- Laboratoire de Spectrométrie de Masse Protéomique, Institute Curie, PSL Research University, Paris, France
| | - Nicolas Molinie
- BIOC, CNRS UMR7654, Institut Polytechnique de Paris, F-91128 Palaiseau, France
| | - Stephane Romero
- BIOC, CNRS UMR7654, Institut Polytechnique de Paris, F-91128 Palaiseau, France
| | - Daniel Bouvard
- INSERM, Institut Albert Bonniot U823, F-38042 Grenoble, France
| | - Daniel Louvard
- CNRS, UMR144, Institute Curie, PSL Research University, F-75005 Paris, France
| | - Alexis M Gautreau
- BIOC, CNRS UMR7654, Institut Polytechnique de Paris, F-91128 Palaiseau, France
| | - Eric Pasmant
- Institut Cochin, INSERM U1016, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Dominique Lallemand
- CNRS, UMR144, Institute Curie, PSL Research University, F-75005 Paris, France.
| |
Collapse
|
273
|
Joukov V, De Nicolo A. The Centrosome and the Primary Cilium: The Yin and Yang of a Hybrid Organelle. Cells 2019; 8:E701. [PMID: 31295970 PMCID: PMC6678760 DOI: 10.3390/cells8070701] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 12/27/2022] Open
Abstract
Centrosomes and primary cilia are usually considered as distinct organelles, although both are assembled with the same evolutionary conserved, microtubule-based templates, the centrioles. Centrosomes serve as major microtubule- and actin cytoskeleton-organizing centers and are involved in a variety of intracellular processes, whereas primary cilia receive and transduce environmental signals to elicit cellular and organismal responses. Understanding the functional relationship between centrosomes and primary cilia is important because defects in both structures have been implicated in various diseases, including cancer. Here, we discuss evidence that the animal centrosome evolved, with the transition to complex multicellularity, as a hybrid organelle comprised of the two distinct, but intertwined, structural-functional modules: the centriole/primary cilium module and the pericentriolar material/centrosome module. The evolution of the former module may have been caused by the expanding cellular diversification and intercommunication, whereas that of the latter module may have been driven by the increasing complexity of mitosis and the requirement for maintaining cell polarity, individuation, and adhesion. Through its unique ability to serve both as a plasma membrane-associated primary cilium organizer and a juxtanuclear microtubule-organizing center, the animal centrosome has become an ideal integrator of extracellular and intracellular signals with the cytoskeleton and a switch between the non-cell autonomous and the cell-autonomous signaling modes. In light of this hypothesis, we discuss centrosome dynamics during cell proliferation, migration, and differentiation and propose a model of centrosome-driven microtubule assembly in mitotic and interphase cells. In addition, we outline the evolutionary benefits of the animal centrosome and highlight the hierarchy and modularity of the centrosome biogenesis networks.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, 197758 Saint-Petersburg, Russia.
| | | |
Collapse
|
274
|
Luqman A, Ebner P, Reichert S, Sass P, Kabagema-Bilan C, Heilmann C, Ruth P, Götz F. A new host cell internalisation pathway for SadA-expressing staphylococci triggered by excreted neurochemicals. Cell Microbiol 2019; 21:e13044. [PMID: 31099148 PMCID: PMC6771854 DOI: 10.1111/cmi.13044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
Abstract
Staphylococcus aureus is a facultative intracellular pathogen that invades a wide range of professional and nonprofessional phagocytes by triggering internalisation by interaction of surface‐bound adhesins with corresponding host cell receptors. Here, we identified a new concept of host cell internalisation in animal‐pathogenic staphylococcal species. This new mechanism exemplified by Staphylococcus pseudintermedius ED99 is not based on surface‐bound adhesins but is due to excreted small neurochemical compounds, such as trace amines (TAs), dopamine (DOP), and serotonin (SER), that render host cells competent for bacterial internalisation. The neurochemicals are produced by only one enzyme, the staphylococcal aromatic amino acid decarboxylase (SadA). Here, we unravelled the mechanism of how neurochemicals trigger internalisation into the human colon cell line HT‐29. We found that TAs and DOP are agonists of the α2‐adrenergic receptor, which, when activated, induces a cascade of reactions involving a decrease in the cytoplasmic cAMP level and an increase in F‐actin formation. The signalling cascade of SER follows a different pathway. SER interacts with 5HT receptors that trigger F‐actin formation without decreasing the cytoplasmic cAMP level. The neurochemical‐induced internalisation in host cells is independent of the fibronectin‐binding protein pathway and has an additive effect. In a sadA deletion mutant, ED99ΔsadA, internalisation was decreased approximately threefold compared with that of the parent strain, and treating S. aureus USA300 with TAs increased internalisation by approximately threefold.
Collapse
Affiliation(s)
- Arif Luqman
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany.,Biology Department, Institut Teknologi Sepuluh Nopember, Surabaya, Indonesia.,Microbiology Division, Generasi Biologi Indonesia (Genbinesia) Foundation, Gresik, Indonesia
| | - Patrick Ebner
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Sebastian Reichert
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | - Peter Sass
- Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| | | | - Christine Heilmann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Peter Ruth
- Institute for Pharmacy, University of Tübingen, Tübingen, Germany
| | - Friedrich Götz
- Microbial Genetics, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Tübingen, Germany
| |
Collapse
|
275
|
Dawson TR, Wente SR. The Plk1 Piece of the Nuclear Envelope Disassembly Puzzle. Dev Cell 2019; 43:115-117. [PMID: 29065301 DOI: 10.1016/j.devcel.2017.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Reporting in this issue of Developmental Cell, Linder et al. (2017) and Martino et al. (2017) reveal in highly complementary studies that Plk1 is recruited to the nuclear pore complex upon mitotic entry, where it acts with Cdk1 to hyperphosphorylate nucleoporin interfaces to promote NPC disassembly and nuclear envelope breakdown.
Collapse
Affiliation(s)
- T Renee Dawson
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Susan R Wente
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
276
|
Zou Z, Gan S, Liu S, Li R, Huang J. Investigation of differentially expressed genes in nasopharyngeal carcinoma by integrated bioinformatics analysis. Oncol Lett 2019; 18:916-926. [PMID: 31289570 DOI: 10.3892/ol.2019.10382] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 04/10/2019] [Indexed: 12/20/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a common malignancy of the head and neck. The aim of the present study was to conduct an integrated bioinformatics analysis of differentially expressed genes (DEGs) and to explore the molecular mechanisms of NPC. Two profiling datasets, GSE12452 and GSE34573, were downloaded from the Gene Expression Omnibus database and included 44 NPC specimens and 13 normal nasopharyngeal tissues. R software was used to identify the DEGs between NPC and normal nasopharyngeal tissues. Distributions of DEGs in chromosomes were explored based on the annotation file and the CYTOBAND database of DAVID. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were applied. Additionally, a protein-protein interaction (PPI) network, constructed using the STRING database and visualized by Cytoscape, was used to identify hub genes, key modules and important transcription factors (TFs). A total of 906 DEGs were identified; 434 (47.90%) DEGs were upregulated and 472 (52.10%) were downregulated. The DEGs were demonstrated to be enriched in chromosome 7p15-p14, 2q31, 1q21-q22, 1q21, 4q21 and 1p31-p22. DEGs were mainly enriched for the following GO terms: 'Cilium movement', 'microtubule bundle formation' and 'axoneme assembly'. KEGG pathway enrichment analysis revealed that pathways for 'cell cycle', 'DNA replication', 'interleukin-17 signaling', 'amoebiasis' and 'glutathione metabolism' were enriched. In addition, a PPI network comprising 867 nodes and 1,241 edges was constructed. Finally, five hub genes (aurora kinase A, cell division cycle 6, mitotic arrest deficient 2-like 1, DNA topoisomerase 2α and TPX2 microtubule nucleation factor), 8 modules, and 14 TFs were identified. Modules analysis revealed that cyclin-dependent kinase 1 and exportin 1 were involved in the pathway of Epstein-Barr virus infection. In summary, the hub genes, key modules and TFs identified in this study may promote our understanding of the pathogenesis of NPC and require further in-depth investigation.
Collapse
Affiliation(s)
- Zhenning Zou
- Department of Pathology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Siyuan Gan
- Department of Pathology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
| | - Rujia Li
- Department of Pathology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Jian Huang
- Department of Pathology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
277
|
Thammahong A, Dhingra S, Bultman KM, Kerkaert JD, Cramer RA. An Ssd1 Homolog Impacts Trehalose and Chitin Biosynthesis and Contributes to Virulence in Aspergillus fumigatus. mSphere 2019; 4:e00244-19. [PMID: 31068436 PMCID: PMC6506620 DOI: 10.1128/msphere.00244-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022] Open
Abstract
Regulation of fungal cell wall biosynthesis is critical to maintain cell wall integrity in dynamic fungal infection microenvironments. Genes involved in this response that impact fungal fitness and host immune responses remain to be fully defined. In this study, we observed that a yeast ssd1 homolog, ssdA, in the filamentous fungus Aspergillus fumigatus is involved in trehalose and cell wall homeostasis. An ssdA null mutant strain exhibited an increase in trehalose levels and a reduction in fungal colony growth rate. In contrast, overexpression of ssdA perturbed trehalose biosynthesis and reduced germination of conidia. The ssdA null mutant strain was more resistant to cell wall-perturbing agents, while overexpression of ssdA increased sensitivity. Overexpression of ssdA significantly increased chitin levels, and both loss and overexpression of ssdA altered subcellular localization of the class V chitin synthase CsmA. Strikingly, overexpression of ssdA abolished adherence to abiotic surfaces and severely attenuated the virulence of A. fumigatus in a murine model of invasive pulmonary aspergillosis. Despite the severe in vitro fitness defects observed upon loss of ssdA, neither surface adherence nor murine survival was impacted. In conclusion, A. fumigatus SsdA plays a critical role in cell wall homeostasis impacting A. fumigatus-host interactions.IMPORTANCE The incidence of life-threatening infections caused by the filamentous fungus Aspergillus fumigatus is increasing along with an increase in the number of fungal strains resistant to contemporary antifungal therapies. The fungal cell wall and the associated carbohydrates required for its synthesis and maintenance are attractive drug targets given that many genes encoding proteins involved in cell wall biosynthesis and integrity are absent in humans. Importantly, genes and associated cell wall biosynthesis and homeostasis regulatory pathways remain to be fully defined in A. fumigatus In this report, we identify SsdA as an important component of trehalose and fungal cell wall biosynthesis in A. fumigatus that consequently impacts the host immune response and fungal virulence in animal models of infection.
Collapse
Affiliation(s)
- Arsa Thammahong
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Sourabh Dhingra
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Katherine M Bultman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Joshua D Kerkaert
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
278
|
Miettinen TP, Kang JH, Yang LF, Manalis SR. Mammalian cell growth dynamics in mitosis. eLife 2019; 8:44700. [PMID: 31063131 PMCID: PMC6534395 DOI: 10.7554/elife.44700] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 05/05/2019] [Indexed: 12/20/2022] Open
Abstract
The extent and dynamics of animal cell biomass accumulation during mitosis are unknown, primarily because growth has not been quantified with sufficient precision and temporal resolution. Using the suspended microchannel resonator and protein synthesis assays, we quantify mass accumulation and translation rates between mitotic stages on a single-cell level. For various animal cell types, growth rates in prophase are commensurate with or higher than interphase growth rates. Growth is only stopped as cells approach metaphase-to-anaphase transition and growth resumes in late cytokinesis. Mitotic arrests stop growth independently of arresting mechanism. For mouse lymphoblast cells, growth in prophase is promoted by CDK1 through increased phosphorylation of 4E-BP1 and cap-dependent protein synthesis. Inhibition of CDK1-driven mitotic translation reduces daughter cell growth. Overall, our measurements counter the traditional dogma that growth during mitosis is negligible and provide insight into antimitotic cancer chemotherapies.
Collapse
Affiliation(s)
- Teemu P Miettinen
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Joon Ho Kang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States.,Department of Physics, Massachusetts Institute of Technology, Cambridge, United States
| | - Lucy F Yang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Scott R Manalis
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
279
|
Expression of CPEB1 gene affects the cycle of ovarian granulosa cells from adult and young goats. ELECTRON J BIOTECHN 2019. [DOI: 10.1016/j.ejbt.2019.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
280
|
Daldello EM, Luong XG, Yang CR, Kuhn J, Conti M. Cyclin B2 is required for progression through meiosis in mouse oocytes. Development 2019; 146:dev172734. [PMID: 30952665 PMCID: PMC6503990 DOI: 10.1242/dev.172734] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/01/2019] [Indexed: 12/20/2022]
Abstract
Cyclins associate with cyclin-dependent serine/threonine kinase 1 (CDK1) to generate the M phase-promoting factor (MPF) activity essential for progression through mitosis and meiosis. Although cyclin B1 (CCNB1) is required for embryo development, previous studies concluded that CCNB2 is dispensable for cell cycle progression. Given previous findings of high Ccnb2 mRNA translation rates in prophase-arrested oocytes, we re-evaluated the role of this cyclin during meiosis. Ccnb2-/- oocytes underwent delayed germinal vesicle breakdown and showed defects during the metaphase-to-anaphase transition. This defective maturation was associated with compromised Ccnb1 and Moloney sarcoma oncogene (Mos) mRNA translation, delayed spindle assembly and increased errors in chromosome segregation. Given these defects, a significant percentage of oocytes failed to complete meiosis I because the spindle assembly checkpoint remained active and anaphase-promoting complex/cyclosome function was inhibited. In vivo, CCNB2 depletion caused ovulation of immature oocytes, premature ovarian failure, and compromised female fecundity. These findings demonstrate that CCNB2 is required to assemble sufficient pre-MPF for timely meiosis re-entry and progression. Although endogenous cyclins cannot compensate, overexpression of CCNB1/2 rescues the meiotic phenotypes, indicating similar molecular properties but divergent modes of regulation of these cyclins.
Collapse
Affiliation(s)
- Enrico Maria Daldello
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Xuan G Luong
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Cai-Rong Yang
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Jonathan Kuhn
- Cell and Tissue Biology Department, University of California, San Francisco, CA 94143, USA
| | - Marco Conti
- Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA 94143, USA
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
281
|
Self-Organized Nuclear Positioning Synchronizes the Cell Cycle in Drosophila Embryos. Cell 2019; 177:925-941.e17. [PMID: 30982601 DOI: 10.1016/j.cell.2019.03.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/16/2018] [Accepted: 03/04/2019] [Indexed: 11/22/2022]
Abstract
The synchronous cleavage divisions of early embryogenesis require coordination of the cell-cycle oscillator, the dynamics of the cytoskeleton, and the cytoplasm. Yet, it remains unclear how spatially restricted biochemical signals are integrated with physical properties of the embryo to generate collective dynamics. Here, we show that synchronization of the cell cycle in Drosophila embryos requires accurate nuclear positioning, which is regulated by the cell-cycle oscillator through cortical contractility and cytoplasmic flows. We demonstrate that biochemical oscillations are initiated by local Cdk1 inactivation and spread through the activity of phosphatase PP1 to generate cortical myosin II gradients. These gradients cause cortical and cytoplasmic flows that control proper nuclear positioning. Perturbations of PP1 activity and optogenetic manipulations of cortical actomyosin disrupt nuclear spreading, resulting in loss of cell-cycle synchrony. We conclude that mitotic synchrony is established by a self-organized mechanism that integrates the cell-cycle oscillator and embryo mechanics.
Collapse
|
282
|
Al Jord A, Spassky N, Meunier A. Motile ciliogenesis and the mitotic prism. Biol Cell 2019; 111:199-212. [PMID: 30905068 DOI: 10.1111/boc.201800072] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022]
Abstract
Motile cilia of epithelial multiciliated cells transport vital fluids along organ lumens to promote essential respiratory, reproductive and brain functions. Progenitors of multiciliated cells undergo massive and coordinated organelle remodelling during their differentiation for subsequent motile ciliogenesis. Defects in multiciliated cell differentiation lead to severe cilia-related diseases by perturbing cilia-based flows. Recent work designated the machinery of mitosis as the orchestrator of the orderly progression of differentiation associated with multiple motile cilia formation. By examining the events leading to motile ciliogenesis with a methodological prism of mitosis, we contextualise and discuss the recent findings to broaden the spectrum of questions related to the differentiation of mammalian multiciliated cells.
Collapse
Affiliation(s)
- Adel Al Jord
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS 7241 INSERM U1050, PSL Research University, Paris, 75005, France
| | - Nathalie Spassky
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris, F-75005, France.,CNRS, UMR 8197, Paris, F-75005, France.,INSERM, U1024, Paris, F-75005, France
| | - Alice Meunier
- Institut de Biologie de l'École Normale Supérieure (IBENS), Paris Sciences et Lettres (PSL) Research University, Paris, F-75005, France.,CNRS, UMR 8197, Paris, F-75005, France.,INSERM, U1024, Paris, F-75005, France
| |
Collapse
|
283
|
Martins GB, Giacomelli G, Goldbeck O, Seibold GM, Bramkamp M. Substrate-dependent cluster density dynamics of Corynebacterium glutamicum phosphotransferase system permeases. Mol Microbiol 2019; 111:1335-1354. [PMID: 30748039 PMCID: PMC6850760 DOI: 10.1111/mmi.14224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2019] [Indexed: 11/29/2022]
Abstract
Many bacteria take up carbohydrates by membrane‐integral sugar specific phosphoenolpyruvate‐dependent carbohydrate:phosphotransferase systems (PTS). Although the PTS is centrally involved in regulation of carbon metabolism in different bacteria, little is known about localization and putative oligomerization of the permease subunits (EII). Here, we analyzed localization of the fructose specific PtsF and the glucose specific PtsG transporters, as well as the general components EI and HPr from Corynebacterium glutamicum using widefield and single molecule localization microscopy. PtsF and PtsG form membrane embedded clusters that localize in a punctate pattern. Size, number and fluorescence of the membrane clusters change upon presence or absence of the transported substrate, and a direct influence of EI and HPr was not observed. In presence of the transport substrate, EII clusters significantly increased in size. Photo‐activated localization microscopy data revealed that, in presence of different carbon sources, the number of EII proteins per cluster remains the same, however, the density of these clusters reduces. Our work reveals a simple mechanism for efficient membrane occupancy regulation. Clusters of PTS EII transporters are densely packed in absence of a suitable substrate. In presence of a transported substrate, the EII proteins in individual clusters occupy larger membrane areas.
Collapse
Affiliation(s)
- Gustavo Benevides Martins
- Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2-4, Planegg-Martinsried, 82152, Germany
| | - Giacomo Giacomelli
- Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2-4, Planegg-Martinsried, 82152, Germany
| | - Oliver Goldbeck
- Institute of Microbiology and Biotechnology, Ulm University, Albert-Einstein Allee 11, Ulm, 89081, Germany
| | - Gerd M Seibold
- Institute of Microbiology and Biotechnology, Ulm University, Albert-Einstein Allee 11, Ulm, 89081, Germany
| | - Marc Bramkamp
- Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2-4, Planegg-Martinsried, 82152, Germany
| |
Collapse
|
284
|
Hasan AA, von Websky K, Reichetzeder C, Tsuprykov O, Gaballa MMS, Guo J, Zeng S, Delić D, Tammen H, Klein T, Kleuser B, Hocher B. Mechanisms of GLP-1 receptor-independent renoprotective effects of the dipeptidyl peptidase type 4 inhibitor linagliptin in GLP-1 receptor knockout mice with 5/6 nephrectomy. Kidney Int 2019; 95:1373-1388. [PMID: 30979564 DOI: 10.1016/j.kint.2019.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 01/14/2019] [Accepted: 01/17/2019] [Indexed: 01/01/2023]
Abstract
Dipeptidyl peptidase type 4 (DPP-4) inhibitors were reported to have beneficial effects in experimental models of chronic kidney disease. The underlying mechanisms are not completely understood. However, these effects could be mediated via the glucagon-like peptide-1 (GLP-1)/GLP-1 receptor (GLP1R) pathway. Here we investigated the renal effects of the DPP-4 inhibitor linagliptin in Glp1r-/- knock out and wild-type mice with 5/6 nephrectomy (5/6Nx). Mice were allocated to groups: sham+wild type+placebo; 5/6Nx+ wild type+placebo; 5/6Nx+wild type+linagliptin; sham+knock out+placebo; 5/6Nx+knock out+ placebo; 5/6Nx+knock out+linagliptin. 5/6Nx caused the development of renal interstitial fibrosis, significantly increased plasma cystatin C and creatinine levels and suppressed renal gelatinase/collagenase, matrix metalloproteinase-1 and -13 activities; effects counteracted by linagliptin treatment in wildtype and Glp1r-/- mice. Two hundred ninety-eight proteomics signals were differentially regulated in kidneys among the groups, with 150 signals specific to linagliptin treatment as shown by mass spectrometry. Treatment significantly upregulated three peptides derived from collagen alpha-1(I), thymosin β4 and heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) and significantly downregulated one peptide derived from Y box binding protein-1 (YB-1). The proteomics results were further confirmed using western blot and immunofluorescence microscopy. Also, 5/6Nx led to significant up-regulation of renal transforming growth factor-β1 and pSMAD3 expression in wild type mice and linagliptin significantly counteracted this up-regulation in wild type and Glp1r-/- mice. Thus, the renoprotective effects of linagliptin cannot solely be attributed to the GLP-1/GLP1R pathway, highlighting the importance of other signaling pathways (collagen I homeostasis, HNRNPA1, YB-1, thymosin β4 and TGF-β1) influenced by DPP-4 inhibition.
Collapse
Affiliation(s)
- Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany; Institute of Nutritional Sciences, University of Potsdam, Potsdam, Germany; Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt; UP Transfer GmbH, University of Potsdam, Potsdam, Germany
| | - Karoline von Websky
- Institute of Nutritional Sciences, University of Potsdam, Potsdam, Germany; Center for Cardiovascular Research, Charité, Berlin, Germany
| | - Christoph Reichetzeder
- Institute of Nutritional Sciences, University of Potsdam, Potsdam, Germany; UP Transfer GmbH, University of Potsdam, Potsdam, Germany; Center for Cardiovascular Research, Charité, Berlin, Germany
| | - Oleg Tsuprykov
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany; Center for Cardiovascular Research, Charité, Berlin, Germany; IFLB GmbH, Institute for Laboratory Medicine, Berlin, Germany
| | - Mohamed M S Gaballa
- Institute of Nutritional Sciences, University of Potsdam, Potsdam, Germany; Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Jingli Guo
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Shufei Zeng
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Denis Delić
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Burkhard Kleuser
- Institute of Nutritional Sciences, University of Potsdam, Potsdam, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany; LADR GmbH Neuruppin MVZ, Neuruppin, Germany; Department of Basic Medicine, Medical college of Hunan Normal University, Changsha, China.
| |
Collapse
|
285
|
Bae H, Go YH, Kwon T, Sung BJ, Cha HJ. A Theoretical Model for the Cell Cycle and Drug Induced Cell Cycle Arrest of FUCCI Systems with Cell-to-Cell Variation during Mitosis. Pharm Res 2019; 36:57. [DOI: 10.1007/s11095-019-2570-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/11/2019] [Indexed: 12/16/2022]
|
286
|
Yi ZY, Liang QX, Meng TG, Li J, Dong MZ, Hou Y, Ouyang YC, Zhang CH, Schatten H, Sun QY, Qiao J, Qian WP. PKCβ1 regulates meiotic cell cycle in mouse oocyte. Cell Cycle 2019; 18:395-412. [PMID: 30730241 DOI: 10.1080/15384101.2018.1564492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PKCβI, a member of the classical protein kinase C family, plays key roles in regulating cell cycle transition. Here, we report the expression, localization and functions of PKCβI in mouse oocyte meiotic maturation. PKCβI and p-PKCβI (phosphor-PKCβI) were expressed from germinal vesicle (GV) stage to metaphase II (MII) stage. Confocal microscopy revealed that PKCβI was localized in the GV and evenly distributed in the cytoplasm after GV breakdown (GVBD), and it was concentrated at the midbody at telophase in meiotic oocytes. While, p-PKCβI was concentrated at the spindle poles at the metaphase stages and associated with midbody at telophase. Depletion of PKCβI by specific siRNA injection resulted in defective spindles, accompanied with spindle assembly checkpoint activation, metaphase I arrest and failure of first polar body (PB1) extrusion. Live cell imaging analysis also revealed that knockdown of PKCβI resulted in abnormal spindles, misaligned chromosomes, and meiotic arrest of oocytes arrest at the Pro-MI/MI stage. PKCβI depletion did not affect the G2/M transition, but its overexpression delayed the G2/M transition through regulating Cyclin B1 level and Cdc2 activity. Our findings reveal that PKCβI is a critical regulator of meiotic cell cycle progression in oocytes. Abbreviations: PKC, protein kinase C; COC, cumulus-oocyte complexes; GV, germinal vesicle; GVBD, germinal vesicle breakdown; Pro-MI, first pro-metaphase; MI, first metaphase; Tel I, telophase I; MII, second metaphase; PB1, first polar body; SAC, spindle assembly checkpoint.
Collapse
Affiliation(s)
- Zi-Yun Yi
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Qiu-Xia Liang
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Tie-Gang Meng
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Jian Li
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Ming-Zhe Dong
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Yi Hou
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Ying-Chun Ouyang
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Chun-Hui Zhang
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| | - Heide Schatten
- c Department of Veterinary Pathobiology , University of Missouri-Columbia , Columbia , MO , USA
| | - Qing-Yuan Sun
- b State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences , Beijing , China
| | - Jie Qiao
- d Reproductive Medical Center , Peking University Third Hospital , Beijing , China
| | - Wei-Ping Qian
- a The Reproductive Medicine Center , Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center , Shenzhen , China
| |
Collapse
|
287
|
Levasseur MD, Thomas C, Davies OR, Higgins JMG, Madgwick S. Aneuploidy in Oocytes Is Prevented by Sustained CDK1 Activity through Degron Masking in Cyclin B1. Dev Cell 2019; 48:672-684.e5. [PMID: 30745144 PMCID: PMC6416240 DOI: 10.1016/j.devcel.2019.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 10/22/2018] [Accepted: 12/29/2018] [Indexed: 01/10/2023]
Abstract
Successful mitosis requires that cyclin B1:CDK1 kinase activity remains high until chromosomes are correctly aligned on the mitotic spindle. It has therefore been unclear why, in mammalian oocyte meiosis, cyclin B1 destruction begins before chromosome alignment is complete. Here, we resolve this paradox and show that mouse oocytes exploit an imbalance in the ratio of cyclin B1 to CDK1 to control CDK1 activity; early cyclin B1 destruction reflects the loss of an excess of non-CDK1-bound cyclin B1 in late prometaphase, while CDK1-bound cyclin B1 is destroyed only during metaphase. The ordered destruction of the two forms of cyclin B1 is brought about by a previously unidentified motif that is accessible in free cyclin B1 but masked when cyclin B1 is in complex with CDK1. This protects the CDK1-bound fraction from destruction in prometaphase, ensuring a period of prolonged CDK1 activity sufficient to achieve optimal chromosome alignment and prevent aneuploidy. In mouse oocytes, an excess of cyclin B1 preserves CDK1 activity A motif in non-CDK1-bound cyclin B1 confers preferential APC/C targeting Non-CDK1-bound cyclin B1 is gradually destroyed before CDK1-bound cyclin B1 Prolonged CDK1 activity assists the spindle checkpoint and prevents aneuploidy
Collapse
Affiliation(s)
- Mark D Levasseur
- Cell Division Biology Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Christopher Thomas
- Cell Division Biology Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Owen R Davies
- Cell Division Biology Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Jonathan M G Higgins
- Cell Division Biology Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Suzanne Madgwick
- Cell Division Biology Group, Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
288
|
Alkhouli MF, Hung J, Squire M, Anderson M, Castro M, Babu JR, Al-Nakkash L, Broderick TL, Plochocki JH. Exercise and resveratrol increase fracture resistance in the 3xTg-AD mouse model of Alzheimer's disease. Altern Ther Health Med 2019; 19:39. [PMID: 30717730 PMCID: PMC6360737 DOI: 10.1186/s12906-019-2451-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/29/2019] [Indexed: 12/18/2022]
Abstract
Background Alzheimer’s disease (AD) and osteoporosis are progressive diseases that affect the elderly population. Both conditions are associated with fracture risk that is greater than twice that of the healthy population. Resveratrol and exercise are two treatments that have been linked with attenuation of age-related diseases, including the risk of bone fractures. In this study, we test the hypothesis that these treatments improve fracture resistance in a mouse model representative of the AD condition. Methods Three-month-old male 3xTg-AD mice were treated for 4 months with resveratrol or exercise or both combined, and compared with wild type mice. Exercise training was performed on a treadmill at 15 m/min for 45 min/day, 5 days/week. Resveratrol was given at 4 g/kg diet in the form of pellets. Three-point bending, cross-sectional geometric, and fluorescence analyses were conducted on tibias and compared by treatment group. Results Tibias of 3xTg mice exhibited signs of diminished bone quality and fracture under less force than age-matched wild type mice (P < 0.05). Treatment with both resveratrol and exercise improved indicators of fracture resistance and bone quality in AD mice to levels comparable to that of wild type mice (P < 0.05). Conclusions The 3xTg mouse model of AD is at elevated risk for limb bone fracture compared to wild type controls. Treatment with resveratrol, exercise, or both in combination improves fracture resistance and cross-sectional geometric indicators of bone strength.
Collapse
|
289
|
Salahuddin S, Fath EK, Biel N, Ray A, Moss CR, Patel A, Patel S, Hilding L, Varn M, Ross T, Cramblet WT, Lowrey A, Pagano JS, Shackelford J, Bentz GL. Epstein-Barr Virus Latent Membrane Protein-1 Induces the Expression of SUMO-1 and SUMO-2/3 in LMP1-positive Lymphomas and Cells. Sci Rep 2019; 9:208. [PMID: 30659232 PMCID: PMC6338769 DOI: 10.1038/s41598-018-36312-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/12/2018] [Indexed: 11/17/2022] Open
Abstract
Epstein-Barr Virus latent membrane protein-1 (LMP1) interacts with the SUMO-conjugating enzyme Ubc9, which induces protein sumoylation and may contribute to LMP1-mediated oncogenesis. After analyzing human lymphoma tissues and EBV-positive cell lines, we now document a strong correlation between LMP1 and sumo-1/2/3 or SUMO-1/2/3 levels, and show that LMP1-induced sumo expression requires the activation of NF-κB signaling through CTAR1 and CTAR2. Together, these results point to a second mechanism by which LMP1 dysregulates sumoylation processes and adds EBV-associated lymphomas to the list of malignancies associated with increased SUMO expression.
Collapse
Affiliation(s)
- Sadia Salahuddin
- Departments of Medicine and Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA.,Atta-ur-Rehman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Emma K Fath
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA
| | - Natalie Biel
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Ashley Ray
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - C Randall Moss
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Akash Patel
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Sheetal Patel
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Leslie Hilding
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Matthew Varn
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Tabithia Ross
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Wyatt T Cramblet
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Angela Lowrey
- Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA
| | - Joseph S Pagano
- Departments of Medicine and Microbiology and Immunology, The University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA
| | - Julia Shackelford
- Department of Cellular Biology and Physiology, The University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA
| | - Gretchen L Bentz
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC, USA. .,Department of Basic Medical Sciences, Mercer University School of Medicine, Macon, GA, USA.
| |
Collapse
|
290
|
Aktar K, Kafi A, Dahiya R. Association of Gpx1 fluctuation in cell cycle progression. In Vitro Cell Dev Biol Anim 2019; 55:94-103. [PMID: 30632027 DOI: 10.1007/s11626-018-00314-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 12/09/2018] [Indexed: 01/19/2023]
Abstract
This research demonstrates fluctuation of glutathione peroxidase1 (Gpx1) throughout cell cycle progression with significant decreased expression at mitosis of HeLa cell. This was achieved with western blot (WB) analysis of target proteins from each phase of synchronized cells. The synchronizations were performed with double thymidine (T/T) for G1/S arrest and thymidine followed by nocodazole (T/N) for G2/M arrest. The G1/S arrested cells were released in fresh medium for 3, 6, 9, 10, and 15h to obtain cell at each phase such as gap1 (G1), synthesis (S), gap2 (G2), mitosis (M), and gap1 (G1) phase, respectively, for investigating Gpx1 expression throughout a complete cycle. The synchronizations were confirmed using fluorescence activated cell sorting (FACS) and WB analysis of phase-specific markers. The fluctuations of Gpx1 expression were verified with universal protein actin and peroxiredoxin1 (Prx1) which are stable throughout the cell cycle. Intriguingly, immunoblots showed the level of Gpx1 decreases at mitosis phase and increased during mitotic exit to G1 phase in HeLa cells, while Prx1 protein level remained constant. The fractionation experiments reveal that only the cytosolic Gpx1 was decreased while their levels at mitochondria remain constant. The highest levels of mitochondrial ROS were measured in mitosis phase with FACS analysis using Mito sox indicating that antioxidant activity of Gpx1 for detoxifying excessive induced endogenous reactive oxygen species (ROS) in the mitosis phase could be the reason for such decreasing level. For unfolding the molecular mechanism of such decreased expression, the Gpx1 was investigated at transcriptional, translational, and proteosomal level. The results revealed that translational mechanism is involve in the decreased expression rather than transcriptional or proteosomal degradation at mitosis phase. This finding supports that Gpx1 is involved in the cell cycle progression through regulation of endogenous ROS. Based on this observation, further research could uncover their possible association with the infinitive division of a cancer cell.
Collapse
Affiliation(s)
- Khudishta Aktar
- BEST group, School of Engineering, University of Glasgow, Glasgow, UK
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Abdul Kafi
- BEST group, School of Engineering, University of Glasgow, Glasgow, UK
| | - Ravinder Dahiya
- BEST group, School of Engineering, University of Glasgow, Glasgow, UK.
| |
Collapse
|
291
|
Okamoto S, Asgar NF, Yokota S, Saito K, Minokoshi Y. Role of the α2 subunit of AMP-activated protein kinase and its nuclear localization in mitochondria and energy metabolism-related gene expressions in C2C12 cells. Metabolism 2019; 90:52-68. [PMID: 30359677 DOI: 10.1016/j.metabol.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK), a heterotrimer with α1 or α2 catalytic subunits, acts as an energy sensor and regulates cellular homeostasis. Whereas AMPKα1 is necessary for myogenesis in skeletal muscle, the role of AMPKα2 in myogenic differentiation and energy metabolism-related gene expressions has remained unclear. We here examined the specific roles of AMPKα1 and AMPKα2 in the myogenic differentiation and mitochondria and energy metabolism-related gene expressions in C2C12 cells. MATERIALS AND METHODS Stable C2C12 cell lines expressing a scramble short hairpin RNA (shRNA) or shRNAs specific for AMPKα1 (shAMPKα1), AMPKα2 (shAMPKα2), or both AMPKα1 and AMPKα2 (shPanAMPK) were generated by lentivirus infection. Lentiviruses encoding wild-type AMPKα2 (WT-AMPKα2) or AMPKα2 with a mutated nuclear localization signal (ΔNLS-AMPKα2) were also constructed for introduction into myoblasts. Myogenesis was induced by culture of C2C12 myoblasts for 6 days in differentiation medium. RESULTS The amount of AMPKα2 increased progressively, whereas that of AMPKα1 remained constant, during the differentiation of myoblasts into myotubes. Expression of shPanAMPK or shAMPKα1, but not that of shAMPKα2, attenuated the proliferation of myoblasts as well as the phosphorylation of both acetyl-CoA carboxylase and the autophagy-initiating kinase ULK1 in myotubes. Up-regulation of myogenin mRNA, a marker for the middle stage of myogenesis, was attenuated in differentiating myotubes expressing shPanAMPK or shAMPKα1. In contrast, up-regulation of gene expression for muscle creatine kinase (MCK), a late-stage differentiation marker, as well as for genes related to mitochondrial biogenesis including the transcriptional coactivator peroxisome proliferator-activated receptor-γ coactivator-1α1 and α4 (PGC-1α1 and PGC-1α4) and mitochondria-specific genes such as cytochrome c were attenuated in myotubes expressing shAMPKα2 or shPanAMPK. The diameter of myotubes expressing shPanAMPK or shAMPKα2 was reduced, whereas that of those expressing shAMPKα1 was increased, compared with myotubes expressing scramble shRNA. A portion of AMPKα2 became localized to the nucleus during myogenic differentiation. The AMPK activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) and 2-deoxyglucose (2DG) each induced the nuclear translocation of WT-AMPKα2, but not that of ΔNLS-AMPKα2. Finally, expression of WT-AMPKα2 increased the mRNA abundance of PGC-1α1 and MCK mRNAs as well as cell diameter and tended to increase that of PGC-1α4, whereas that of ΔNLS-AMPKα2 increased only the abundance of MCK mRNA, in myotubes depleted of endogenous AMPKα2. CONCLUSION TAMPKα1 and AMPKα2 have distinct roles in myogenic differentiation of C2C12 cells, with AMPKα1 contributing to the middle stage of myogenesis and AMPKα2 to the late stage. AMPKα2 regulates gene expressions including MCK, PGC-1α1 and PGC-1α4 and mitochondria-specific genes such as cytochrome c during the late stage of differentiation. Furthermore, the nuclear translocation of AMPKα2 is necessary for maintenance of PGC-1α1 mRNA during myogenesis.
Collapse
Affiliation(s)
- Shiki Okamoto
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Second Department of Internal Medicine (Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology), Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Nur Farehan Asgar
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Shigefumi Yokota
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Kumiko Saito
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
292
|
Farshadi E, Yan J, Leclere P, Goldbeter A, Chaves I, van der Horst GTJ. The positive circadian regulators CLOCK and BMAL1 control G2/M cell cycle transition through Cyclin B1. Cell Cycle 2019; 18:16-33. [PMID: 30558467 PMCID: PMC6343743 DOI: 10.1080/15384101.2018.1558638] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 01/09/2023] Open
Abstract
We previously identified a tight bidirectional phase coupling between the circadian clock and the cell cycle. To understand the role of the CLOCK/BMAL1 complex, representing the main positive regulator of the circadian oscillator, we knocked down Bmal1 or Clock in NIH3T33C mouse fibroblasts (carrying fluorescent reporters for clock and cell cycle phase) and analyzed timing of cell division in individual cells and cell populations. Inactivation of Bmal1 resulted in a loss of circadian rhythmicity and a lengthening of the cell cycle, originating from delayed G2/M transition. Subsequent molecular analysis revealed reduced levels of Cyclin B1, an important G2/M regulator, upon suppression of Bmal1 gene expression. In complete agreement with these experimental observations, simulation of Bmal1 knockdown in a computational model for coupled mammalian circadian clock and cell cycle oscillators (now incorporating Cyclin B1 induction by BMAL1) revealed a lengthening of the cell cycle. Similar data were obtained upon knockdown of Clock gene expression. In conclusion, the CLOCK/BMAL1 complex controls cell cycle progression at the level of G2/M transition through regulation of Cyclin B1 expression.
Collapse
Affiliation(s)
- Elham Farshadi
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jie Yan
- Unit of Theoretical Chronobiology, Faculty of Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pierre Leclere
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Albert Goldbeter
- Unit of Theoretical Chronobiology, Faculty of Sciences, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Inês Chaves
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|
293
|
Janks L, Sprague RS, Egan TM. ATP-Gated P2X7 Receptors Require Chloride Channels To Promote Inflammation in Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2018; 202:883-898. [PMID: 30598517 DOI: 10.4049/jimmunol.1801101] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 11/25/2018] [Indexed: 12/28/2022]
Abstract
Immune cells of myeloid origin show robust expression of ATP-gated P2X7 receptors, two-transmembrane ion channels permeable to Na+, K+, and Ca2+ Receptor activation promotes inflammasome activation and release of the proinflammatory cytokines IL-1β and IL-18. In this study, we show that ATP generates facilitating cationic currents in monocyte-derived human macrophages and permeabilizes the plasma membrane to polyatomic cationic dyes. We find that antagonists of PLA2 and Cl- channels abolish P2X7 receptor-mediated current facilitation, membrane permeabilization, blebbing, phospholipid scrambling, inflammasome activation, and IL-1β release. Our data demonstrate significant differences in the actions of ATP in murine and human macrophages and suggest that PLA2 and Cl- channels mediate innate immunity downstream of P2X7 receptors in human macrophages.
Collapse
Affiliation(s)
- Laura Janks
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Randy S Sprague
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| | - Terrance M Egan
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO 63104
| |
Collapse
|
294
|
Greenwald EC, Mehta S, Zhang J. Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks. Chem Rev 2018; 118:11707-11794. [PMID: 30550275 PMCID: PMC7462118 DOI: 10.1021/acs.chemrev.8b00333] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cellular signaling networks are the foundation which determines the fate and function of cells as they respond to various cues and stimuli. The discovery of fluorescent proteins over 25 years ago enabled the development of a diverse array of genetically encodable fluorescent biosensors that are capable of measuring the spatiotemporal dynamics of signal transduction pathways in live cells. In an effort to encapsulate the breadth over which fluorescent biosensors have expanded, we endeavored to assemble a comprehensive list of published engineered biosensors, and we discuss many of the molecular designs utilized in their development. Then, we review how the high temporal and spatial resolution afforded by fluorescent biosensors has aided our understanding of the spatiotemporal regulation of signaling networks at the cellular and subcellular level. Finally, we highlight some emerging areas of research in both biosensor design and applications that are on the forefront of biosensor development.
Collapse
Affiliation(s)
- Eric C Greenwald
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Sohum Mehta
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| | - Jin Zhang
- University of California , San Diego, 9500 Gilman Drive, BRFII , La Jolla , CA 92093-0702 , United States
| |
Collapse
|
295
|
Al-Essa MK, Melzer S, Tárnok A. Two-Color Analysis of Leukocytes Labeled by Modified RBCs and Their Fragments. Cytometry A 2018; 95:339-346. [PMID: 30578619 DOI: 10.1002/cyto.a.23682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 09/06/2018] [Accepted: 10/25/2018] [Indexed: 11/08/2022]
Abstract
Red blood cells (RBCs) are attractive tools for surface modification to adhere specifically to molecules, cellular fragments (e.g., microvesicles), or whole cells for potential use in bioanalytical assays or as a delivery vehicle in targeted therapy. Within this study, we have loaded RBCs with fluorochrome-conjugated antibodies (Ab) against CD45 and CD22 leukocyte markers and evaluated the conjugation process by microscopy. We have assessed the potential application of RBCs fragments generated from conjugated RBCs for targeting Cyto-Trol control cells by flow cytometric (FCM) approaches. Based on their scattering and fluorescence characteristics (FITC and PE expression), modified RBCs and their fragments, Cyto-Trol cells, and clusters of both were distinguished by two color FCM analysis. Fragments with anti-human Kallestad Ab as a nonspecific FITC conjugate had less than 20% binding to Cyto-Trol controls compared to CD45-FITC Ab conjugate with nearly 100% binding capacity. Cyto-Trol-microvesicle-clusters were more than 45% positive for either FITC or PE. Anti-CD22-PE modified RBCs fragments were also useful in staining and showing about 19.5% positively stained events in the Cyto-Trol region. The proof-of-concept shows, that specific antibody can be attached to RBCs, and generated fragments can be useful to stain target cells for FCM analysis. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Mohamed K Al-Essa
- Department of Physiology and Biochemistry, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Susanne Melzer
- Clinical Trial Center Leipzig, Leipzig University, Leipzig, Germany
| | - Attila Tárnok
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany.,Department of Precision Instrument, Tsinghua University, Beijing, China.,Department of Therapy Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| |
Collapse
|
296
|
Ani CJ, Obayemi JD, Uzonwanne VO, Danyuo Y, Odusanya OS, Hu J, Malatesta K, Soboyejo WO. A shear assay study of single normal/breast cancer cell deformation and detachment from poly-di-methyl-siloxane (PDMS) surfaces. J Mech Behav Biomed Mater 2018; 91:76-90. [PMID: 30544025 DOI: 10.1016/j.jmbbm.2018.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/25/2018] [Accepted: 11/12/2018] [Indexed: 02/01/2023]
Abstract
This paper presents the results of a combined experimental and analytical/computational study of viscoelastic cell deformation and detachment from poly-di-methyl-siloxane (PDMS) surfaces. Fluid mechanics and fracture mechanics concepts are used to model the detachment of biological cells observed under shear assay conditions. The analytical and computational models are used to compute crack driving forces, which are then related to crack extension during the detachment of normal breast cells and breast cancer cells from PDMS surfaces that are relevant to biomedical implants. The interactions between cells and the extracellular matrix, or the extracellular matrix and the PDMS substrate, are then characterized using force microscopy measurements of the pull-off forces that are used to determine the adhesion energies. Finally, fluorescence microscopy staining of the cytosketelal structures (actin, micro-tubulin and cyto-keratin), transmembrane proteins (vimentin) and the ECM structures (Arginin Glycine Aspartate - RGD) is used to show that the detachment of cells during the shear assay experiments occurs via interfacial cracking between (between the ECM and the cell membranes) with a high incidence of crack bridging by transmembrane vinculin structures that undergo pull-out until they detach from the actin cytoskeletal structure. The implications of the results are discussed for the design of interfaces that are relevant to implantable biomedical devices and normal/cancer tissue.
Collapse
Affiliation(s)
- C J Ani
- Department of Theoretical and Applied Physics, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria; Department of Physics, Salem University, Km 16, PMB 1060, Lokoja, Kogi State, Nigeria
| | - J D Obayemi
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA
| | - V O Uzonwanne
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA
| | - Y Danyuo
- Department of Mechanical Engineering, Ashesi University, Berekuso, Ghana; Department of Materials Science and Engineering, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria
| | - O S Odusanya
- Advanced Biotechnology Laboratory, Sheda Science and Technology Complex, Abuja, Nigeria
| | - J Hu
- Princeton Institute for the Science and Technology of Materials (PRISM), and The Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - K Malatesta
- Princeton Institute for the Science and Technology of Materials (PRISM), and The Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - W O Soboyejo
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA; Department of Materials Science and Engineering, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria; Advanced Biotechnology Laboratory, Sheda Science and Technology Complex, Abuja, Nigeria.
| |
Collapse
|
297
|
Do AV, Worthington K, Tucker B, Salem AK. Controlled drug delivery from 3D printed two-photon polymerized poly(ethylene glycol) dimethacrylate devices. Int J Pharm 2018; 552:217-224. [PMID: 30268853 PMCID: PMC6204107 DOI: 10.1016/j.ijpharm.2018.09.065] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
Abstract
Controlled drug delivery systems have been utilized to enhance the therapeutic effects of many drugs by delivering drugs in a time-dependent and sustained manner. Here, with the aid of 3D printing technology, drug delivery devices were fabricated and tested using a model drug (fluorophore: rhodamine B). Poly(ethylene glycol) dimethacrylate (PEGDMA) devices were fabricated using a two-photon polymerization (TPP) system and rhodamine B was homogenously entrapped inside the polymer matrix during photopolymerization. These devices were printed with varying porosity and morphology using varying printing parameters such as slicing and hatching distance. The effects of these variables on drug release kinetics were determined by evaluating device fluorescence over the course of one week. These PEGDMA-based structures were then investigated for toxicity and biocompatibility in vitro, where MTS assays were performed using a range of cell types including induced pluripotent stem cells (iPSCs). Overall, tuning the hatching distance, slicing distance, and pore size of the fabricated devices modulated the rhodamine B release profile, in each case presumably due to resulting changes in the motility of the small molecule and its access to structure edges. In general, increased spacing provided higher drug release while smaller spacing resulted in some occlusion, preventing media infiltration and thus resulting in reduced fluorophore release. The devices had no cytotoxic effects on human embryonic kidney cells (HEK293), bone marrow stromal stem cells (BMSCs) or iPSCs. Thus, we have demonstrated the utility of two-photon polymerization to create biocompatible, complex miniature devices with fine details and tunable release of a model drug.
Collapse
Affiliation(s)
- Anh-Vu Do
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa,Department of Chemical and Biochemical Engineering, College of Engineering, The University of Iowa
| | - Kristan Worthington
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, College of Medicine, The University of Iowa,Department of Biomedical Engineering, College of Engineering, The University of Iowa
| | - Budd Tucker
- Institute for Vision Research, Department of Ophthalmology and Visual Sciences, College of Medicine, The University of Iowa
| | - Aliasger K. Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa,Department of Chemical and Biochemical Engineering, College of Engineering, The University of Iowa,Department of Biomedical Engineering, College of Engineering, The University of Iowa,
| |
Collapse
|
298
|
Boonthum C, Namdee K, Khongkow M, Temisak S, Chatdarong K, Sajomsang W, Ponglowhapan S, Yata T. Gonadotropin-releasing hormone-modified chitosan as a safe and efficient gene delivery vector for spermatogonia cells. Reprod Domest Anim 2018; 53 Suppl 3:23-28. [DOI: 10.1111/rda.13346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 09/05/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Chatwalee Boonthum
- Department of Obstetrics, Gynaecology and Reproduction; Research Unit of Obstetrics and Reproduction in Animals; Faculty of Veterinary Science; Chulalongkorn University; Bangkok Thailand
| | - Katawut Namdee
- National Nanotechnology Centre (NANOTEC); National Science and Technology Development Agency; Pathumthani Thailand
| | - Mattaka Khongkow
- National Nanotechnology Centre (NANOTEC); National Science and Technology Development Agency; Pathumthani Thailand
| | - Sasithont Temisak
- Bio Analysis Group, Chemical Metrology and Biometry Department; National Institute of Metrology (NIMT); Pathumthani Thailand
| | - Kaywalee Chatdarong
- Department of Obstetrics, Gynaecology and Reproduction; Research Unit of Obstetrics and Reproduction in Animals; Faculty of Veterinary Science; Chulalongkorn University; Bangkok Thailand
| | - Warayuth Sajomsang
- National Nanotechnology Centre (NANOTEC); National Science and Technology Development Agency; Pathumthani Thailand
| | - Suppawiwat Ponglowhapan
- Department of Obstetrics, Gynaecology and Reproduction; Research Unit of Obstetrics and Reproduction in Animals; Faculty of Veterinary Science; Chulalongkorn University; Bangkok Thailand
| | - Teerapong Yata
- National Nanotechnology Centre (NANOTEC); National Science and Technology Development Agency; Pathumthani Thailand
| |
Collapse
|
299
|
Tadić V, Adam A, Goldhammer N, Lautenschlaeger J, Oberstadt M, Malci A, Le TT, Sengupta S, Stubendorff B, Keiner S, Witte OW, Grosskreutz J. Investigation of mitochondrial calcium uniporter role in embryonic and adult motor neurons from G93A hSOD1 mice. Neurobiol Aging 2018; 75:209-222. [PMID: 30597405 DOI: 10.1016/j.neurobiolaging.2018.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/16/2018] [Accepted: 11/17/2018] [Indexed: 11/19/2022]
Abstract
Amyotrophic lateral sclerosis is characterized by progressive death of motor neurons (MNs) with glutamate excitotoxicity and mitochondrial Ca2+ overload as critical mechanisms in disease pathophysiology. We used MNs from G93AhSOD1 and nontransgenic embryonic cultures and adult mice to analyze the expression of the main mitochondrial calcium uniporter (MCU). MCU was overexpressed in cultured embryonic G93AhSOD1 MNs compared to nontransgenic MNs but downregulated in MNs from adult G93AhSOD1 mice. Furthermore, cultured embryonic G93AhSOD1 were rescued from kainate-induced excitotoxicity by the Ca2+/calmodulin-dependent protein kinase type II inhibitor; KN-62, which reduced MCU expression in G93AhSOD1 MNs. MCU activation via kaempferol neither altered MCU expression nor influenced MN survival. However, its acute application served as a fine tool to study spontaneous Ca2+ activity in cultured neurons which was significantly altered by the mutated hSOD1. Pharmacological manipulation of MCU expression might open new possibilities to fight excitotoxic damage in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Vedrana Tadić
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany.
| | - Adam Adam
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Nadine Goldhammer
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Janin Lautenschlaeger
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany; Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Moritz Oberstadt
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Ayse Malci
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Thanh Tu Le
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Saikata Sengupta
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | | | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
300
|
Saatian B, Austin RS, Tian G, Chen C, Nguyen V, Kohalmi SE, Geelen D, Cui Y. Analysis of a novel mutant allele of GSL8 reveals its key roles in cytokinesis and symplastic trafficking in Arabidopsis. BMC PLANT BIOLOGY 2018; 18:295. [PMID: 30466394 PMCID: PMC6249969 DOI: 10.1186/s12870-018-1515-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 10/31/2018] [Indexed: 05/10/2023]
Abstract
BACKGROUND Plant cell walls are mainly composed of polysaccharides such as cellulose and callose. Callose exists at a very low level in the cell wall; however, it plays critical roles at different stages of plant development as well as in defence against unfavorable conditions. Callose is accumulated at the cell plate, at plasmodesmata and in male and female gametophytes. Despite the important roles of callose in plants, the mechanisms of its synthesis and regulatory properties are not well understood. RESULTS CALLOSE SYNTHASE (CALS) genes, also known as GLUCAN SYNTHASE-LIKE (GSL), comprise a family of 12 members in Arabidopsis thaliana. Here, we describe a new allele of GSL8 (named essp8) that exhibits pleiotropic seedling defects. Reduction of callose deposition at the cell plates and plasmodesmata in essp8 leads to ectopic endomitosis and an increase in the size exclusion limit of plasmodesmata during early seedling development. Movement of two non-cell-autonomous factors, SHORT ROOT and microRNA165/6, both required for root radial patterning during embryonic root development, are dysregulated in the primary root of essp8. This observation provides evidence for a molecular mechanism explaining the gsl8 root phenotype. We demonstrated that GSL8 interacts with PLASMODESMATA-LOCALIZED PROTEIN 5, a β-1,3-glucanase, and GSL10. We propose that they all might be part of a putative callose synthase complex, allowing a concerted regulation of callose deposition at plasmodesmata. CONCLUSION Analysis of a novel mutant allele of GSL8 reveals that GSL8 is a key player in early seedling development in Arabidopsis. GSL8 is required for maintaining the basic ploidy level and regulating the symplastic trafficking. Callose deposition at plasmodesmata is highly regulated and occurs through interaction of different components, likely to be incorporated into a callose biosynthesis complex. We are providing new evidence supporting an earlier hypothesis that GSL8 might have regulatory roles apart from its enzymatic function in plasmodesmata regulation.
Collapse
Affiliation(s)
- Behnaz Saatian
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
- Department of Biology, Western University, 1391 Sandford St, London, ON N5V 4T3 Canada
| | - Ryan S. Austin
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
- Department of Biology, Western University, 1391 Sandford St, London, ON N5V 4T3 Canada
| | - Gang Tian
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
| | - Chen Chen
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
- Department of Biology, Western University, 1391 Sandford St, London, ON N5V 4T3 Canada
| | - Vi Nguyen
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
| | - Susanne E. Kohalmi
- Department of Biology, Western University, 1391 Sandford St, London, ON N5V 4T3 Canada
| | - Danny Geelen
- In Vitro Biology and Horticulture, Department of Plant Production, University of Ghent, 9000 Ghent, Belgium
| | - Yuhai Cui
- Agriculture and Agri-Food Canada, London Research and Development Centre, London, ON Canada
- Department of Biology, Western University, 1391 Sandford St, London, ON N5V 4T3 Canada
| |
Collapse
|