201
|
Liu S, Pellman D. The coordination of nuclear envelope assembly and chromosome segregation in metazoans. Nucleus 2020; 11:35-52. [PMID: 32208955 PMCID: PMC7289584 DOI: 10.1080/19491034.2020.1742064] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 01/25/2023] Open
Abstract
The nuclear envelope (NE) is composed of two lipid bilayer membranes that enclose the eukaryotic genome. In interphase, the NE is perforated by thousands of nuclear pore complexes (NPCs), which allow transport in and out of the nucleus. During mitosis in metazoans, the NE is broken down and then reassembled in a manner that enables proper chromosome segregation and the formation of a single nucleus in each daughter cell. Defects in coordinating NE reformation and chromosome segregation can cause aberrant nuclear architecture. This includes the formation of micronuclei, which can trigger a catastrophic mutational process commonly observed in cancers called chromothripsis. Here, we discuss the current understanding of the coordination of NE reformation with chromosome segregation during mitotic exit in metazoans. We review differing models in the field and highlight recent work suggesting that normal NE reformation and chromosome segregation are physically linked through the timing of mitotic spindle disassembly.
Collapse
Affiliation(s)
- Shiwei Liu
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Pellman
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
202
|
Zheng R, Shi Z, Li W, Yu J, Wang Y, Zhou Q. Identification and prognostic value of DLGAP5 in endometrial cancer. PeerJ 2020; 8:e10433. [PMID: 33312770 PMCID: PMC7703392 DOI: 10.7717/peerj.10433] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/05/2020] [Indexed: 01/01/2023] Open
Abstract
Background Endometrial cancer poses a serious threat to women’s health worldwide, and its pathogenesis, although actively explored, is not fully understood. DLGAP5 is a recently identified cell cycle-regulation gene not reported in endometrial cancer. This study was aiming to analyze the role of DLGAP5 in tumorigenesis and development and to investigate its prognostic significance of patients with endometrial cancer. Methodology Microarray datasets (GSE17025, GSE39099 and GSE63678) from the GEO database were used for comparative analysis, and their intersection was obtained by applying the Venn diagram, and DLGAP5 was selected as the target gene. Next, transcriptome data (n = 578) was downloaded from TCGA-UCEC to analyze the mRNA expression profile of DLGAP5. Then, immunohistochemical data provided by HPA were used to identify the different protein expression levels of DLGAP5 in tumor tissues and normal tissues. Subsequently, the prognostic meaning of DLGAP5 in patients with endometrial cancer was explored based on survival data from TCGA-UCEC (n = 541). Finally, the reliability of DLGAP5 expression was verified by RT-qPCR. Results Transcriptome data from TCGA-UCEC, immunohistochemical data from HPA, and RT-qPCR results from clinical samples were used for triple validation to confirm that the expression of DLGAP5 in endometrial cancer tissues was significantly higher than that in normal endometrial tissues. Kaplan–Meier survival analysis announced that the expression level of DLGAP5 was negatively correlated with the overall survival of patients with endometrial cancer. Conclusions DLGAP5 is a potential oncogene with cell cycle regulation, and its overexpression can predict the poor prognosis of patients with endometrial cancer. As a candidate target for the diagnosis and treatment of endometrial cancer, it is worthwhile to make further study to reveal the carcinogenicity of DLGAP5 and the mechanism of its resistance of organisms.
Collapse
Affiliation(s)
- Ruoyi Zheng
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengzheng Shi
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenzhi Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jianqin Yu
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuli Wang
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qing Zhou
- Department of Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
203
|
Lin K, Zhu X, Luo C, Bu F, Zhu J, Zhu Z. Data mining combined with experiments to validate CEP55 as a prognostic biomarker in colorectal cancer. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:167-182. [PMID: 33190424 PMCID: PMC7860595 DOI: 10.1002/iid3.375] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is a common tumor with high morbidity and mortality. Current specific diagnosis regarding CRC remains complicated and costly, and specific diagnostic biomarkers are lacking. METHODS To find potential diagnostic and prognostic biomarkers for CRC, we screened and analyzed many CRC sequencing data by The Cancer Genome Atlas Program and Gene Expression Omnibus, and validated that CEP55 may be a potential diagnostic biomarker for CRC by molecular cytological experiments and immunohistochemistry, among others. RESULTS We found that CEP55 is upregulated in CRC tissues and tumor cells and can promote CRC proliferation and metastasis by activating the p53/p21 axis and that CEP55 mutations in tumor patients result in worse overall survival and disease-free survival time. Besides, we also found that genes, such as CDK1, CCNB1, NEK2, KIF14, CDCA5, and RFC3 were upregulated in tumors, and their mutations would affect the prognosis of CRC patients, but these results await for more experimental evidence. CONCLUSION Our study validates CEP55 as a potential diagnostic and prognostic biomarker for CRC, and we also provide multiple genes and potential molecular mechanisms that may serve as diagnostic and prognostic markers for CRC.
Collapse
Affiliation(s)
- Kang Lin
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chen Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Fanqin Bu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinfeng Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhengming Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
204
|
Rosendo-Pineda MJ, Vicente JJ, Vivas O, Pacheco J, Loza-Huerta A, Sampieri A, Wordeman L, Moreno C, Vaca L. Phosphorylation of NMDA receptors by cyclin B/CDK1 modulates calcium dynamics and mitosis. Commun Biol 2020; 3:665. [PMID: 33184446 PMCID: PMC7665045 DOI: 10.1038/s42003-020-01393-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/15/2020] [Indexed: 12/02/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDAR) are glutamate-gated calcium channels named after their artificial agonist. NMDAR are implicated in cell proliferation under normal and pathophysiological conditions. However, the role of NMDAR during mitosis has not yet been explored in individual cells. We found that neurotransmitter-evoked calcium entry via endogenous NMDAR in cortical astrocytes was transient during mitosis. The same occurred in HEK293 cells transfected with the NR1/NR2A subunits of NMDAR. This transient calcium entry during mitosis was due to phosphorylation of the first intracellular loop of NMDAR (S584 of NR1 and S580 of NR2A) by cyclin B/CDK1. Expression of phosphomimetic mutants resulted in transient calcium influx and enhanced NMDAR inactivation independent of the cell cycle phase. Phosphomimetic mutants increased entry of calcium in interphase and generated several alterations during mitosis: increased mitotic index, increased number of cells with lagging chromosomes and fragmentation of pericentriolar material. In summary, by controlling cytosolic calcium, NMDAR modulate mitosis and probably cell differentiation/proliferation. Our results suggest that phosphorylation of NMDAR by cyclin B/CDK1 during mitosis is required to preserve mitotic fidelity. Altering the modulation of the NMDAR by cyclin B/CDK1 may conduct to aneuploidy and cancer.
Collapse
Affiliation(s)
| | - Juan Jesus Vicente
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Jonathan Pacheco
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Arlet Loza-Huerta
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Alicia Sampieri
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Claudia Moreno
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA
| | - Luis Vaca
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, DF, 04510, Mexico.
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
205
|
Abdullah CS, Ray P, Alam S, Kale N, Aishwarya R, Morshed M, Dutta D, Hudziak C, Banerjee SK, Mallik S, Banerjee S, Bhuiyan MS, Quadir M. Chemical Architecture of Block Copolymers Differentially Abrogate Cardiotoxicity and Maintain the Anticancer Efficacy of Doxorubicin. Mol Pharm 2020; 17:4676-4690. [PMID: 33151075 DOI: 10.1021/acs.molpharmaceut.0c00963] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The molecular architecture of pH-responsive amphiphilic block copolymers, their self-assembly behavior to form nanoparticles (NPs), and doxorubicin (DOX)-loading technique govern the extent of DOX-induced cardiotoxicity. We observed that the choice of pH-sensitive tertiary amines, surface charge, and DOX-loading techniques within the self-assembled NPs strongly influence the release and stimulation of DOX-induced cardiotoxicity in primary cardiomyocytes. However, covalent conjugation of DOX to a pH-sensitive nanocarrier through a "conditionally unstable amide" linkage (PCPY-cDOX; PC = polycarbonate and PY = 2-pyrrolidine-1-yl-ethyl-amine) significantly reduced the cardiotoxicity of DOX in cardiomyocytes as compared to noncovalently encapsulated DOX NPs (PCPY-eDOX). When these formulations were tested for drug release in serum-containing media, the PCPY-cDOX systems showed prolonged control over drug release (for ∼72 h) at acidic pH compared to DOX-encapsulated nanocarriers, as expected. We found that DOX-encapsulated nanoformulations triggered cardiotoxicity in primary cardiomyocytes more acutely, while conjugated systems such as PCPY-cDOX prevented cardiotoxicity by disabling the nuclear entry of the drug. Using 2D and 3D (spheroid) cultures of an ER + breast cancer cell line (MCF-7) and a triple-negative breast cancer cell line (MDA-MB-231), we unravel that, similar to encapsulated systems (PCPY-eDOX-type) as reported earlier, the PCPY-cDOX system suppresses cellular proliferation in both cell lines and enhances trafficking through 3D spheroids of MDA-MB-231 cells. Collectively, our studies indicate that PCPY-cDOX is less cardiotoxic as compared to noncovalently encapsulated variants without compromising the chemotherapeutic properties of the drug. Thus, our studies suggest that the appropriate selection of the nanocarrier for DOX delivery may prove fruitful in shifting the balance between low cardiotoxicity and triggering the chemotherapeutic potency of DOX.
Collapse
Affiliation(s)
- Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States
| | - Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States
| | - Narendra Kale
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States
| | - Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Cathleen Hudziak
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Sushanta K Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri 64128, United States.,Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana 71103, United States
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, North Dakota 58108, United States
| |
Collapse
|
206
|
Margarido AS, Bornes L, Vennin C, van Rheenen J. Cellular Plasticity during Metastasis: New Insights Provided by Intravital Microscopy. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a037267. [PMID: 31615867 DOI: 10.1101/cshperspect.a037267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Metastasis is a highly dynamic process during which cancer and microenvironmental cells undergo a cascade of events required for efficient dissemination throughout the body. During the metastatic cascade, tumor cells can change their state and behavior, a phenomenon commonly defined as cellular plasticity. To monitor cellular plasticity during metastasis, high-resolution intravital microscopy (IVM) techniques have been developed and allow us to visualize individual cells by repeated imaging in animal models. In this review, we summarize the latest technological advancements in the field of IVM and how they have been applied to monitor metastatic events. In particular, we highlight how longitudinal imaging in native tissues can provide new insights into the plastic physiological and developmental processes that are hijacked by cancer cells during metastasis.
Collapse
Affiliation(s)
- Andreia S Margarido
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Laura Bornes
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Claire Vennin
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
207
|
Barajas-Martínez A, Bermeo K, de la Cruz L, Martínez-Vargas M, Martínez-Tapia RJ, García DE, Navarro L. Cannabinoid receptors are differentially regulated in the pancreatic islets during the early development of metabolic syndrome. Islets 2020; 12:134-144. [PMID: 33289595 PMCID: PMC7751681 DOI: 10.1080/19382014.2020.1849927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The endocannabinoid system is found in tissues that regulate the glycemia, including adipose tissue, muscle, and pancreatic islets. Diet-induced metabolic syndrome changes the expression of the CB receptors in muscle, adipose tissue, and liver. However, it is poorly understood whether metabolic syndrome (MetS) affects the expression of CB receptors in pancreatic β cells. We analyzed the expression of CB receptors in pancreatic β cells under chronic high-sucrose diet (HSD)-induced MetS. Wistar rats fed an HSD as a model of MetS were used to investigate changes in cannabinoid receptors. After 8 weeks of treatment, we evaluated the appearance of the following MetS biomarkers: glucose intolerance, hyperinsulinemia, insulin resistance, hypertriglyceridemia, and an increase in visceral adiposity. To determine the presence of CB1 and CB2 receptors in pancreatic β cells, immunofluorescence of primary cell cultures and pancreatic sections was performed. For whole-islet quantification of membrane-bound CB1 and CB2 receptors, western-blotting following differential centrifugation was conducted. Our results revealed that an HSD treatment closely mimics the alterations seen in MetS. We observed that in primary cell culture, CB1 and CB2 receptors were expressed at a higher level in pancreatic β cells compared with non-β cells. MetS resulted in a reduction of CB1 in the islet, whereas abundant CB2 was observed after the treatment. CB1 and CB2 receptors are differentially expressed in pancreatic β cells during MetS development.
Collapse
Affiliation(s)
- Antonio Barajas-Martínez
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Karina Bermeo
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Lizbeth de la Cruz
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Marina Martínez-Vargas
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Ricardo Jesús Martínez-Tapia
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - David Erasmo García
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Luz Navarro
- Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
- CONTACT Luz Navarro Departamento de Fisiología, Universidad Nacional Autónoma de México (UNAM), Ciudad de MéxicoC.P. 04510, México
| |
Collapse
|
208
|
Nicolas HA, Bertrand AT, Labib S, Mohamed-Uvaize M, Bolongo PM, Wu WY, Bilińska ZT, Bonne G, Akimenko MA, Tesson F. Protein Kinase C Alpha Cellular Distribution, Activity, and Proximity with Lamin A/C in Striated Muscle Laminopathies. Cells 2020; 9:cells9112388. [PMID: 33142761 PMCID: PMC7693451 DOI: 10.3390/cells9112388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/19/2020] [Accepted: 10/28/2020] [Indexed: 11/24/2022] Open
Abstract
Striated muscle laminopathies are cardiac and skeletal muscle conditions caused by mutations in the lamin A/C gene (LMNA). LMNA codes for the A-type lamins, which are nuclear intermediate filaments that maintain the nuclear structure and nuclear processes such as gene expression. Protein kinase C alpha (PKC-α) interacts with lamin A/C and with several lamin A/C partners involved in striated muscle laminopathies. To determine PKC-α’s involvement in muscular laminopathies, PKC-α’s localization, activation, and interactions with the A-type lamins were examined in various cell types expressing pathogenic lamin A/C mutations. The results showed aberrant nuclear PKC-α cellular distribution in mutant cells compared to WT. PKC-α activation (phos-PKC-α) was decreased or unchanged in the studied cells expressing LMNA mutations, and the activation of its downstream targets, ERK 1/2, paralleled PKC-α activation alteration. Furthermore, the phos-PKC-α-lamin A/C proximity was altered. Overall, the data showed that PKC-α localization, activation, and proximity with lamin A/C were affected by certain pathogenic LMNA mutations, suggesting PKC-α involvement in striated muscle laminopathies.
Collapse
Affiliation(s)
- Hannah A. Nicolas
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Anne T. Bertrand
- Sorbonne Université, Inserm, Centre de Recherche en Myologie, UMRS 974, G.H. Pitié-Salpêtrière, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Sarah Labib
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Musfira Mohamed-Uvaize
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Pierrette M. Bolongo
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
| | - Wen Yu Wu
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Zofia T. Bilińska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Gisèle Bonne
- Sorbonne Université, Inserm, Centre de Recherche en Myologie, UMRS 974, G.H. Pitié-Salpêtrière, 75013 Paris, France; (A.T.B.); (G.B.)
| | - Marie-Andrée Akimenko
- Department of Biology, Faculty of Science, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (H.A.N.); (W.Y.W.); (M.-A.A.)
| | - Frédérique Tesson
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada; (S.L.); (M.M.-U.); (P.M.B.)
- Correspondence: ; Tel.: +1-613-562-5800 (ext. 7370)
| |
Collapse
|
209
|
Markowitsch SD, Schupp P, Lauckner J, Vakhrusheva O, Slade KS, Mager R, Efferth T, Haferkamp A, Juengel E. Artesunate Inhibits Growth of Sunitinib-Resistant Renal Cell Carcinoma Cells through Cell Cycle Arrest and Induction of Ferroptosis. Cancers (Basel) 2020; 12:cancers12113150. [PMID: 33121039 PMCID: PMC7692972 DOI: 10.3390/cancers12113150] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Renal cell carcinoma (RCC) is the most common kidney malignancy. Due to development of therapy resistance, efficacy of conventional drugs such as sunitinib is limited. Artesunate (ART), a drug originating from Traditional Chinese Medicine, has exhibited anti-tumor effects in several non-urologic tumors. ART inhibited growth, reduced metastatic properties, and curtailed metabolism in sunitinib-sensitive and sunitinib–resistant RCC cells. In three of four tested cell lines, ART’s growth inhibitory effects were accompanied by cell cycle arrest and modulation of cell cycle regulating proteins. In a fourth cell line, KTCTL-26, ART evoked ferroptosis, an iron-dependent cell death, and exhibited stronger anti-tumor effects than in the other cell lines. The regulatory protein, p53, was only detectable in the KTCTL-26 cells, possibly making p53 a predictive marker of cancer that may respond better to ART. ART, therefore, may hold promise as an additive therapy option for selected patients with advanced or therapy-resistant RCC. Abstract Although innovative therapeutic concepts have led to better treatment of advanced renal cell carcinoma (RCC), efficacy is still limited due to the tumor developing resistance to applied drugs. Artesunate (ART) has demonstrated anti-tumor effects in different tumor entities. This study was designed to investigate the impact of ART (1–100 µM) on the sunitinib-resistant RCC cell lines, Caki-1, 786-O, KTCTL26, and A-498. Therapy-sensitive (parental) and untreated cells served as controls. ART’s impact on tumor cell growth, proliferation, clonogenic growth, apoptosis, necrosis, ferroptosis, and metabolic activity was evaluated. Cell cycle distribution, the expression of cell cycle regulating proteins, p53, and the occurrence of reactive oxygen species (ROS) were investigated. ART significantly increased cytotoxicity and inhibited proliferation and clonogenic growth in both parental and sunitinib-resistant RCC cells. In Caki-1, 786-O, and A-498 cell lines growth inhibition was associated with G0/G1 phase arrest and distinct modulation of cell cycle regulating proteins. KTCTL-26 cells were mainly affected by ART through ROS generation, ferroptosis, and decreased metabolism. p53 exclusively appeared in the KTCTL-26 cells, indicating that p53 might be predictive for ART-dependent ferroptosis. Thus, ART may hold promise for treating selected patients with advanced and even therapy-resistant RCC.
Collapse
Affiliation(s)
- Sascha D. Markowitsch
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Patricia Schupp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Julia Lauckner
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Olesya Vakhrusheva
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Kimberly S. Slade
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - René Mager
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Thomas Efferth
- Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Staudingerweg 5, 55128 Mainz, Germany;
| | - Axel Haferkamp
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
| | - Eva Juengel
- Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (S.D.M.); (P.S.); (J.L.); (O.V.); (K.S.S.); (R.M.); (A.H.)
- Correspondence: ; Tel.: +49-631-175-433; Fax: +49-6131-174-410
| |
Collapse
|
210
|
Onodera S, Nakamura Y, Azuma T. Gorlin Syndrome: Recent Advances in Genetic Testing and Molecular and Cellular Biological Research. Int J Mol Sci 2020; 21:E7559. [PMID: 33066274 PMCID: PMC7590212 DOI: 10.3390/ijms21207559] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/08/2023] Open
Abstract
Gorlin syndrome is a skeletal disorder caused by a gain of function mutation in Hedgehog (Hh) signaling. The Hh family comprises of many signaling mediators, which, through complex mechanisms, play several important roles in various stages of development. The Hh information pathway is essential for bone tissue development. It is also the major driver gene in the development of basal cell carcinoma and medulloblastoma. In this review, we first present the recent advances in Gorlin syndrome research, in particular, the signaling mediators of the Hh pathway and their functions at the genetic level. Then, we discuss the phenotypes of mutant mice and Hh signaling-related molecules in humans revealed by studies using induced pluripotent stem cells.
Collapse
Affiliation(s)
- Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kandamisaki-cho Chiyoda-ku, Tokyo 101-0061, Japan;
| | - Yuriko Nakamura
- Department of Oral Oncology, Oral and Maxillofacial Surgery, Tokyo Dental College, 5-11-13 Sugano, Ichikawa, Chiba 272-8513, Japan;
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, 2-9-18 Kandamisaki-cho Chiyoda-ku, Tokyo 101-0061, Japan;
| |
Collapse
|
211
|
Monitoring and modeling of lymphocytic leukemia cell bioenergetics reveals decreased ATP synthesis during cell division. Nat Commun 2020; 11:4983. [PMID: 33020492 PMCID: PMC7536222 DOI: 10.1038/s41467-020-18769-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
The energetic demands of a cell are believed to increase during mitosis, but the rates of ATP synthesis and consumption during mitosis have not been quantified. Here, we monitor mitochondrial membrane potential of single lymphocytic leukemia cells and demonstrate that mitochondria hyperpolarize from the G2/M transition until the metaphase-anaphase transition. This hyperpolarization was dependent on cyclin-dependent kinase 1 (CDK1) activity. By using an electrical circuit model of mitochondria, we quantify mitochondrial ATP synthesis rates in mitosis from the single-cell time-dynamics of mitochondrial membrane potential. We find that mitochondrial ATP synthesis decreases by approximately 50% during early mitosis and increases back to G2 levels during cytokinesis. Consistently, ATP levels and ATP synthesis are lower in mitosis than in G2 in synchronized cell populations. Overall, our results provide insights into mitotic bioenergetics and suggest that cell division is not a highly energy demanding process. ATP drives most cellular processes, although ATP production and consumption levels during mitosis remain unreported. Here, the authors combine metabolic measurements and modeling to quantify ATP levels and synthesis dynamics, revealing that ATP synthesis and consumption are lowered during mitosis.
Collapse
|
212
|
Activation and degranulation of CAR-T cells using engineered antigen-presenting cell surfaces. PLoS One 2020; 15:e0238819. [PMID: 32976541 PMCID: PMC7518621 DOI: 10.1371/journal.pone.0238819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/24/2020] [Indexed: 11/19/2022] Open
Abstract
Adoptive cell transfer of Chimeric Antigen Receptor (CAR)-T cells showed promising results in patients with B cell malignancies. However, the detailed mechanism of CAR-T cell interaction with the target tumor cells is still not well understood. This work provides a systematic method for analyzing the activation and degranulation of second-generation CAR-T cells utilizing antigen-presenting cell surfaces. Antigen-presenting cell surfaces composed of circular micropatterns of CAR-specific anti-idiotype antibodies have been developed to mimic the interaction of CAR-T cells with target tumor cells using micro-contact printing. The levels of activation and degranulation of fixed non-transduced T cells (NT), CD19.CAR-T cells, and GD2.CAR-T cells on the antigen-presenting cell surfaces were quantified and compared by measuring the intensity of the CD3ζ chain phosphorylation and the Lysosome-Associated Membrane Protein 1 (LAMP-1), respectively. The size and morphology of the cells were also measured. The intracellular Ca2+ flux of NT and CAR-T cells upon engagement with the antigen-presenting cell surface was reported. Results suggest that NT and CD19.CAR-T cells have comparable activation levels, while NT have higher degranulation levels than CD19.CAR-T cells and GD2.CAR-T cells. The findings show that antigen-presenting cell surfaces allow a quantitative analysis of the molecules involved in synapse formation in different CAR-T cells in a systematic, reproducible manner.
Collapse
|
213
|
Yano S, Tazawa H, Kagawa S, Fujiwara T, Hoffman RM. FUCCI Real-Time Cell-Cycle Imaging as a Guide for Designing Improved Cancer Therapy: A Review of Innovative Strategies to Target Quiescent Chemo-Resistant Cancer Cells. Cancers (Basel) 2020; 12:cancers12092655. [PMID: 32957652 PMCID: PMC7563319 DOI: 10.3390/cancers12092655] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 01/06/2023] Open
Abstract
Simple Summary Chemotherapy of solid tumors has made very slow progress over many decades. A major problem has been that solid tumors very often contain non-dividing cells due to lack of oxygen deep in the tumor and these non-dividing cells resist most currently-used chemotherapy which usually only targets dividing cells. The present review demonstrates how a unique imaging system, FUCCI, which color codes cells depending on whether they are in a dividing or non-dividing phase, is being used to design very novel therapy that targets non-dividing cancer cells which can greatly improve the efficacy of cancer chemotherapy. Abstract Progress in chemotherapy of solid cancer has been tragically slow due, in large part, to the chemoresistance of quiescent cancer cells in tumors. The fluorescence ubiquitination cell-cycle indicator (FUCCI) was developed in 2008 by Miyawaki et al., which color-codes the phases of the cell cycle in real-time. FUCCI utilizes genes linked to different color fluorescent reporters that are only expressed in specific phases of the cell cycle and can, thereby, image the phases of the cell cycle in real-time. Intravital real-time FUCCI imaging within tumors has demonstrated that an established tumor comprises a majority of quiescent cancer cells and a minor population of cycling cancer cells located at the tumor surface or in proximity to tumor blood vessels. In contrast to most cycling cancer cells, quiescent cancer cells are resistant to cytotoxic chemotherapy, most of which target cells in S/G2/M phases. The quiescent cancer cells can re-enter the cell cycle after surviving treatment, which suggests the reason why most cytotoxic chemotherapy is often ineffective for solid cancers. Thus, quiescent cancer cells are a major impediment to effective cancer therapy. FUCCI imaging can be used to effectively target quiescent cancer cells within tumors. For example, we review how FUCCI imaging can help to identify cell-cycle-specific therapeutics that comprise decoy of quiescent cancer cells from G1 phase to cycling phases, trapping the cancer cells in S/G2 phase where cancer cells are mostly sensitive to cytotoxic chemotherapy and eradicating the cancer cells with cytotoxic chemotherapy most active against S/G2 phase cells. FUCCI can readily image cell-cycle dynamics at the single cell level in real-time in vitro and in vivo. Therefore, visualizing cell cycle dynamics within tumors with FUCCI can provide a guide for many strategies to improve cell-cycle targeting therapy for solid cancers.
Collapse
Affiliation(s)
- Shuya Yano
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Center for Graduate Medical Education, Okayama University Hospital, Okayama 700-8558, Japan
- Correspondence: ; Tel.: +81-86-235-7257; Fax: +81-86-221-8775
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Center of Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (H.T.); (S.K.); (T.F.)
| | - Robert M. Hoffman
- AntiCancer, Inc., San Diego, CA 92111, USA;
- Department of Surgery, University of California, San Diego, CA 92093, USA
| |
Collapse
|
214
|
Lv S, Wang X, Bai X, Ning H, Li Y, Wen H, Lu W, Wang J. Mesenchymal epithelial transition factor regulates tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells through down-regulation of cyclin B1. Int J Biochem Cell Biol 2020; 128:105844. [PMID: 32882404 DOI: 10.1016/j.biocel.2020.105844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 11/25/2022]
Abstract
Tumor necrosis factor-related apoptotic induction ligand can induce cell apoptosis in various tumor cells. However, many cancer cells are resistant to tumor necrosis factor-related apoptotic induction ligand. Therefore, overcoming the tumor necrosis factor-related apoptotic induction ligand resistance makes it possible for tumor necrosis factor-related apoptotic induction ligand-based anti-cancer therapies. In this study, we took mesenchymal epithelial transition factor as the research target to study its role in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma. Mesenchymal epithelial transition factor gene has been proved to be an effective predictor of recurrence after hepatocellular carcinoma resection. The expression of mesenchymal epithelial transition factor and cyclin B1 were measured in tumor necrosis factor-related apoptotic induction ligand-resistant and non-resistant hepatocellular carcinoma tissues. Cyclin B1-knockdown and cyclin B1-overexpression hepatocellular carcinoma cells were treated with tumor necrosis factor-related apoptotic induction ligand; mesenchymal epithelial transition factor knockout, mesenchymal epithelial transition factor re-introduction and cyclin B1 restored in hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand were established. And MTT, bromodeoxyuridine, flow cytometry and western blotting were performed to evaluate the effect of mesenchymal epithelial transition factor and cyclin B1 on hepatocellular carcinoma cells treated with tumor necrosis factor-related apoptotic induction ligand. In addition, subcutaneous tumor transplantation in nude mice was conducted to access the effect of mesenchymal epithelial transition factor and cyclin B1 on tumor formation in vivo. In conclusion, cyclin B1 enhanced the cell growth and inhibited apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells. And mesenchymal epithelial transition factor promoted the cell growth and apoptosis in tumor necrosis factor-related apoptotic induction ligand-resistant hepatocellular carcinoma cells by regulating cyclin B1. Therefore, mesenchymal epithelial transition factor regulates the cyclin B1 to regulate tumor necrosis factor-related apoptotic induction ligand resistance in hepatocellular carcinoma cells. Our results suggest a novel molecular mechanism for regulating tumor necrosis factor-related apoptotic induction ligand resistance, which might be helpful to select drug targets in the treatment of liver cancer.
Collapse
Affiliation(s)
- Shuai Lv
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China.
| | - Xijuan Wang
- Department of Pediatrics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, China
| | - Xia Bai
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hanbing Ning
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Yingxia Li
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Hongtao Wen
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Wenquan Lu
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| | - Jingyun Wang
- Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450018, Henan Province, China
| |
Collapse
|
215
|
Lu S, Sung T, Amaro M, Hirakawa B, Jessen B, Hu W. Phenotypic Characterization of Targeted Knockdown of Cyclin-Dependent Kinases in the Intestinal Epithelial Cells. Toxicol Sci 2020; 177:226-234. [PMID: 32556214 DOI: 10.1093/toxsci/kfaa092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are serine/threonine kinases that regulate cell cycle and have been vigorously pursued as druggable targets for cancer. There are over 20 members of the CDK family. Given their structural similarity, selective inhibition by small molecules has been elusive. In addition, collateral damage to highly proliferative normal cells by CDK inhibitors remains a safety concern. Intestinal epithelial cells are highly proliferative and the impact of individual CDK inhibition on intestinal cell proliferation has not been well studied. Using the rat intestinal epithelial (IEC6) cells as an in vitro model, we found that the selective CDK4/6 inhibitor palbociclib lacked potent anti-proliferative activity in IEC6 relative to the breast cancer cell line MCF7, indicating the absence of intestinal cell reliance on CDK4/6 for cell cycle progression. To further illustrate the role of CDKs in intestinal cells, we chose common targets of CDK inhibitors (CDK 1, 2, 4, 6, and 9) for targeted gene knockdown to evaluate phenotypes. Surprisingly, only CDK1 and CDK9 knockdown demonstrated profound cell death or had moderate growth effects, respectively. CDK2, 4, or 6 knockdowns, whether single, double, or triple combinations, did not have substantial impact. Studies evaluating CDK1 knockdown under various cell seeding densities indicate direct effects on viability independent of proliferation state and imply a potential noncanonical role for CDK1 in intestinal epithelial biology. This research supports the concept that CDK1 and CDK9, but not CDKs 2, 4, or 6, are essential for intestinal cell cycle progression and provides safety confidence for interphase CDK inhibition.
Collapse
Affiliation(s)
- Shuyan Lu
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| | - Tae Sung
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| | - Marina Amaro
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| | - Brad Hirakawa
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| | - Bart Jessen
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| | - Wenyue Hu
- Drug Safety Research and Development, Pfizer Inc., San Diego, California 92121
| |
Collapse
|
216
|
Kara N, Kent MR, Didiano D, Rajaram K, Zhao A, Summerbell ER, Patton JG. The miR-216a-Dot1l Regulatory Axis Is Necessary and Sufficient for Müller Glia Reprogramming during Retina Regeneration. Cell Rep 2020; 28:2037-2047.e4. [PMID: 31433981 PMCID: PMC6750267 DOI: 10.1016/j.celrep.2019.07.061] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 06/13/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
Unlike the adult mammalian retina, Müller glia (MG) in the adult zebrafish retina are able to dedifferentiate into a ‘‘stem cell’’-like state and give rise to multipotent progenitor cells upon retinal damage. We show that miR-216a is downregulated in MG after constant intense light lesioning and that miR-216a suppression is necessary and sufficient for MG dedifferentiation and proliferation during retina regeneration. miR-216a targets the H3K79 methyltransferase Dot1l, which is upregulated in proliferating MG after retinal damage. Loss-of-function experiments show that Dot1l is necessary for MG reprogramming and mediates MG proliferation downstream of miR-216a. We further demonstrate that miR-216a and Dot1l regulate MG-mediated retina regeneration through canonical Wnt signaling. This article reports a regulatory mechanism upstream of Wnt signaling during retina regeneration and provides potential targets for enhancing regeneration in the adult mammalian retina. Unlike the adult mammalian retina, Müller glia in the adult zebrafish retina are able to reprogram into a stem cell-like state and give rise to multipotent progenitor cells upon retinal damage. Kara et al. show that miR-216a suppression stimulates Müller glia reprogramming through upregulation of the H3K79 methyltransferase Dot1l and activation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Nergis Kara
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Matthew R Kent
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Dominic Didiano
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Kamya Rajaram
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Anna Zhao
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Emily R Summerbell
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
217
|
Olbrich T, Vega-Sendino M, Murga M, de Carcer G, Malumbres M, Ortega S, Ruiz S, Fernandez-Capetillo O. A Chemical Screen Identifies Compounds Capable of Selecting for Haploidy in Mammalian Cells. Cell Rep 2020; 28:597-604.e4. [PMID: 31315040 PMCID: PMC6656781 DOI: 10.1016/j.celrep.2019.06.060] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/24/2019] [Accepted: 06/15/2019] [Indexed: 12/24/2022] Open
Abstract
The recent availability of somatic haploid cell lines has provided a unique tool for genetic studies in mammals. However, the percentage of haploid cells rapidly decreases in these cell lines, which we recently showed is due to their overgrowth by diploid cells present in the cultures. Based on this property, we have now performed a phenotypic chemical screen in human haploid HAP1 cells aiming to identify compounds that facilitate the maintenance of haploid cells. Our top hit was 10-Deacetyl-baccatin-III (DAB), a chemical precursor in the synthesis of Taxol, which selects for haploid cells in HAP1 and mouse haploid embryonic stem cultures. Interestingly, DAB also enriches for diploid cells in mixed cultures of diploid and tetraploid cells, including in the colon cancer cell line DLD-1, revealing a general strategy for selecting cells with lower ploidy in mixed populations of mammalian cells. Mammalian haploid cell cultures become progressively enriched in diploid cells DAB, a precursor of Taxol, facilitates the maintenance of haploidy DAB selects for cells with lower ploidy in mixed cultures of mammalian cells Statins accelerate the gradual loss of haploid cells in culture
Collapse
Affiliation(s)
- Teresa Olbrich
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Maria Vega-Sendino
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Matilde Murga
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Guillermo de Carcer
- Chromosome Dynamics Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Marcos Malumbres
- Chromosome Dynamics Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Sagrario Ortega
- Transgenics Unit, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Sergio Ruiz
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Oscar Fernandez-Capetillo
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain; Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 21 Stockholm, Sweden.
| |
Collapse
|
218
|
Djeghloul D, Patel B, Kramer H, Dimond A, Whilding C, Brown K, Kohler AC, Feytout A, Veland N, Elliott J, Bharat TAM, Tarafder AK, Löwe J, Ng BL, Guo Y, Guy J, Huseyin MK, Klose RJ, Merkenschlager M, Fisher AG. Identifying proteins bound to native mitotic ESC chromosomes reveals chromatin repressors are important for compaction. Nat Commun 2020; 11:4118. [PMID: 32807789 PMCID: PMC7431861 DOI: 10.1038/s41467-020-17823-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Epigenetic information is transmitted from mother to daughter cells through mitosis. Here, to identify factors that might play a role in conveying epigenetic memory through cell division, we report on the isolation of unfixed, native chromosomes from metaphase-arrested cells using flow cytometry and perform LC-MS/MS to identify chromosome-bound proteins. A quantitative proteomic comparison between metaphase-arrested cell lysates and chromosome-sorted samples reveals a cohort of proteins that were significantly enriched on mitotic ESC chromosomes. These include pluripotency-associated transcription factors, repressive chromatin-modifiers such as PRC2 and DNA methyl-transferases, and proteins governing chromosome architecture. Deletion of PRC2, Dnmt1/3a/3b or Mecp2 in ESCs leads to an increase in the size of individual mitotic chromosomes, consistent with de-condensation. Similar results were obtained by the experimental cleavage of cohesin. Thus, we identify chromosome-bound factors in pluripotent stem cells during mitosis and reveal that PRC2, DNA methylation and Mecp2 are required to maintain chromosome compaction.
Collapse
Affiliation(s)
- Dounia Djeghloul
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Bhavik Patel
- Flow Cytometry Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Holger Kramer
- Biological Mass Spectrometry and Proteomics Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Andrew Dimond
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Chad Whilding
- Microscopy Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Karen Brown
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Anne-Céline Kohler
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Amelie Feytout
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Nicolas Veland
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - James Elliott
- Flow Cytometry Facility, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Tanmay A M Bharat
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Abul K Tarafder
- Sir William Dunn School of Pathology, University of Oxford, OX1 3RE, Oxford, UK
| | - Jan Löwe
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Bee L Ng
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Ya Guo
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Jacky Guy
- The Wellcome Centre for Cell Biology, University of Edinburgh, Edinburgh, EH9 3BH, UK
| | - Miles K Huseyin
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK
| | - Robert J Klose
- Department of Biochemistry, University of Oxford, OX1 3QU, Oxford, UK
| | - Matthias Merkenschlager
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Amanda G Fisher
- Lymphocyte Development Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
219
|
Naderi R, Saadati Mollaei H, Elofsson A, Hosseini Ashtiani S. Using Micro- and Macro-Level Network Metrics Unveils Top Communicative Gene Modules in Psoriasis. Genes (Basel) 2020; 11:genes11080914. [PMID: 32785106 PMCID: PMC7464240 DOI: 10.3390/genes11080914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 11/22/2022] Open
Abstract
(1) Background: Psoriasis is a multifactorial chronic inflammatory disorder of the skin, with significant morbidity, characterized by hyperproliferation of the epidermis. Even though psoriasis’ etiology is not fully understood, it is believed to be multifactorial, with numerous key components. (2) Methods: In order to cast light on the complex molecular interactions in psoriasis vulgaris at both protein–protein interactions and transcriptomics levels, we studied a set of microarray gene expression analyses consisting of 170 paired lesional and non-lesional samples. Afterwards, a network analysis was conducted on the protein–protein interaction network of differentially expressed genes based on micro- and macro-level network metrics at a systemic level standpoint. (3) Results: We found 17 top communicative genes, all of which were experimentally proven to be pivotal in psoriasis, which were identified in two modules, namely the cell cycle and immune system. Intra- and inter-gene interaction subnetworks from the top communicative genes might provide further insight into the corresponding characteristic interactions. (4) Conclusions: Potential gene combinations for therapeutic/diagnostics purposes were identified. Moreover, our proposed workflow could be of interest to a broader range of future biological network analysis studies.
Collapse
Affiliation(s)
- Reyhaneh Naderi
- Department of Artificial Intelligence and Robotics, Faculty of Computer Engineering, Iran University of Science and Technology, Tehran 1684613114, Iran;
| | - Homa Saadati Mollaei
- Department of Advanced Sciences and Technology, Islamic Azad University Tehran Medical Sciences, Tehran 1916893813, Iran;
| | - Arne Elofsson
- Department of Biochemistry and Biophysics and Science for Life Laboratory, Stockholm University, 106 91 Stockholm, Sweden;
| | - Saman Hosseini Ashtiani
- Department of Biochemistry and Biophysics and Science for Life Laboratory, Stockholm University, 106 91 Stockholm, Sweden;
- Correspondence: ; Tel.: +46-762623644
| |
Collapse
|
220
|
Taubenberger AV, Baum B, Matthews HK. The Mechanics of Mitotic Cell Rounding. Front Cell Dev Biol 2020; 8:687. [PMID: 32850812 PMCID: PMC7423972 DOI: 10.3389/fcell.2020.00687] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/06/2020] [Indexed: 12/21/2022] Open
Abstract
When animal cells enter mitosis, they round up to become spherical. This shape change is accompanied by changes in mechanical properties. Multiple studies using different measurement methods have revealed that cell surface tension, intracellular pressure and cortical stiffness increase upon entry into mitosis. These cell-scale, biophysical changes are driven by alterations in the composition and architecture of the contractile acto-myosin cortex together with osmotic swelling and enable a mitotic cell to exert force against the environment. When the ability of cells to round is limited, for example by physical confinement, cells suffer severe defects in spindle assembly and cell division. The requirement to push against the environment to create space for spindle formation is especially important for cells dividing in tissues. Here we summarize the evidence and the tools used to show that cells exert rounding forces in mitosis in vitro and in vivo, review the molecular basis for this force generation and discuss its function for ensuring successful cell division in single cells and for cells dividing in normal or diseased tissues.
Collapse
Affiliation(s)
- Anna V. Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Buzz Baum
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Helen K. Matthews
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
221
|
Marwah M, Narain Srivastava P, Mishra S, Nagarsenker M. Functionally engineered 'hepato-liposomes': Combating liver-stage malaria in a single prophylactic dose. Int J Pharm 2020; 587:119710. [PMID: 32739383 DOI: 10.1016/j.ijpharm.2020.119710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 01/09/2023]
Abstract
Primaquine continues to remain the gold standard molecule with an incumbent toxicity profile, as far as radical treatment of malaria is concerned. Better molecules are available at experimental level but their targeted delivery is a challenge. The present work identifies 'Decoquinate (DQN)' as a repurposed, safer drug molecule with a potential to function as an appealing replacement for primaquine active against liver-stage malaria. The work focuses on delivering the highly lipophilic DQN (log P ~ 5) in a liposomal carrier system to 'sporozoite infested hepatocytes' using two different in-house synthesized hepatotropic ligands. Functionally engineered 'hepato-liposomes' exhibit differences in their DQN loading capacities but no significant change in morphology or particle size and are also not affected by freeze drying. Two ligands, targeting different receptors on hepatocytes, have been compared for their in vitro and in vivo drug delivery efficiency in liver stage malaria. The studies reveal superior antimalarial efficacy of differently designed DQN loaded liposomes and demonstrate antimalarial efficacy at a low dose of 0.5 mg/kg for a repurposed molecule like DQN. The in vivo studies successfully discriminate the functional efficiency of the carriers and establish the importance of design in liposomal drug delivery for malarial prophylaxis.
Collapse
Affiliation(s)
- Megha Marwah
- Department of Pharmaceutics, Bombay College of Pharmacy, Kalina, Santacruz (East), Mumbai, India
| | - Pratik Narain Srivastava
- Division Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Satish Mishra
- Division Molecular Parasitology and Immunology, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Mangal Nagarsenker
- Department of Pharmaceutics, Bombay College of Pharmacy, Kalina, Santacruz (East), Mumbai, India.
| |
Collapse
|
222
|
Parrilla I, Martinez EA, Gil MA, Cuello C, Roca J, Rodriguez-Martinez H, Martinez CA. Boar seminal plasma: current insights on its potential role for assisted reproductive technologies in swine. Anim Reprod 2020; 17:e20200022. [PMID: 33029213 PMCID: PMC7534575 DOI: 10.1590/1984-3143-ar2020-0022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Seminal plasma (SP) supports not only sperm function but also the ability of spermatozoa to withstand biotechnological procedures as artificial insemination, freezing or sex sorting. Moreover, evidence has been provided that SP contains identifiable molecules which can act as fertility biomarkers, and even improve the output of assisted reproductive technologies by acting as modulators of endometrial and embryonic changes of gene expression, thus affecting embryo development and fertility beyond the sperm horizon. In this overview, we discuss current knowledge of the composition of SP, mainly proteins and cytokines, and their influence on semen basic procedures, such as liquid storage or cryopreservation. The role of SP as modulator of endometrial and embryonic molecular changes that lead to successful pregnancy will also be discussed.
Collapse
Affiliation(s)
- Inmaculada Parrilla
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Emilio Arsenio Martinez
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Maria Antonia Gil
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Cristina Cuello
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Jordi Roca
- Departmento de Medicina y Cirugía Animal, Facultad de Veterinaria, Campus de de Excelencia International "Campus Mare Nostrum", Universidad de Murcia, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria, Campus de Ciencias de la Salud, Murcia, Spain
| | - Heriberto Rodriguez-Martinez
- Department of Biomedical & Clinical Sciences, BKH/Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Cristina Alicia Martinez
- Department of Biomedical & Clinical Sciences, BKH/Obstetrics & Gynaecology, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
223
|
Fatima I, Singh AB, Dhawan P. MASTL: A novel therapeutic target for Cancer Malignancy. Cancer Med 2020; 9:6322-6329. [PMID: 32692487 PMCID: PMC7476815 DOI: 10.1002/cam4.3141] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/09/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting mitotic kinases is an emerging anticancer approach with promising preclinical outcomes. Microtubule‐associated serine/threonine kinase like (MASTL), also known as Greatwall (Gwl), is an important mitotic kinase that regulates mitotic progression of normal or transformed cells by blocking the activity of tumor suppressor protein phosphatase 2A (PP2A). MASTL upregulation has now been detected in multiple cancer types and associated with aggressive clinicopathological features. Apart, an aberrant MASTL activity has been implicated in oncogenic transformation through the development of chromosomal instability and alteration of key oncogenic signaling pathways. In this regard, recent publications have revealed potential role of MASTL in the regulation of AKT/mTOR and Wnt/β‐catenin signaling pathways, which may be independent of its regulation of PP2A‐B55 (PP2A holoenzyme containing a B55‐family regulatory subunit). Taken together, MASTL kinase has emerged as a novel target for cancer therapeutics, and hence development of small molecule inhibitors of MASTL may significantly improve the clinical outcomes of cancer patients. In this article, we review the role of MASTL in cancer progression and the current gaps in this knowledge. We also discuss potential efficacy of MASTL expression for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Iram Fatima
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Amar B Singh
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Punita Dhawan
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
224
|
Munro T, Miller CM, Antunes E, Sharma D. Interactions of Osteoprogenitor Cells with a Novel Zirconia Implant Surface. J Funct Biomater 2020; 11:E50. [PMID: 32708701 PMCID: PMC7565437 DOI: 10.3390/jfb11030050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 02/03/2023] Open
Abstract
Background: This study compared the in vitro response of a mouse pre-osteoblast cell line on a novel sandblasted zirconia surface with that of titanium. Material and Methods: The MC3T3-E1 subclone 4 osteoblast precursor cell line was cultured on either sandblasted titanium (SBCpTi) or sandblasted zirconia (SBY-TZP). The surface topography was analysed by three-dimensional laser microscopy and scanning electron microscope. The wettability of the discs was also assessed. The cellular response was quantified by assessing the morphology (day 1), proliferation (day 1, 3, 5, 7, 9), viability (day 1, 9), and migration (0, 6, 24 h) assays. Results: The sandblasting surface treatment in both titanium and zirconia increased the surface roughness by rendering a defined surface topography with titanium showing more apparent nano-topography. The wettability of the two surfaces showed no significant difference. The zirconia surface resulted in improved cellular spreading and a significantly increased rate of migration compared to titanium. However, the cellular proliferation and viability noted in our experiments were not significantly different on the zirconia and titanium surfaces. Conclusions: The novel, roughened zirconia surface elicited cellular responses comparable to, or exceeding that, of titanium. Therefore, this novel zirconia surface may be an acceptable substitute for titanium as a dental implant material.
Collapse
Affiliation(s)
- Thomas Munro
- College of Medicine and Dentistry, James Cook University, 14-88 McGregor Road, Smithfield 4878, QLD, Australia;
| | - Catherine M. Miller
- College of Public Health, Medical and Veterinary Sciences, James Cook University, 14-88 McGregor Road, Smithfield 4878, QLD, Australia;
- The Australian Institute of Tropical Health and Medicine (AITHM) James Cook University, 14-88 McGregor Road, Smithfield 4878, QLD, Australia
| | - Elsa Antunes
- College of Science & Engineering, James Cook University, 1 James Cook Drive, Douglas, Townsville 4814, QLD, Australia;
| | - Dileep Sharma
- College of Medicine and Dentistry, James Cook University, 14-88 McGregor Road, Smithfield 4878, QLD, Australia;
- The Australian Institute of Tropical Health and Medicine (AITHM) James Cook University, 14-88 McGregor Road, Smithfield 4878, QLD, Australia
| |
Collapse
|
225
|
Xiang Y, Tanaka Y, Patterson B, Hwang SM, Hysolli E, Cakir B, Kim KY, Wang W, Kang YJ, Clement EM, Zhong M, Lee SH, Cho YS, Patra P, Sullivan GJ, Weissman SM, Park IH. Dysregulation of BRD4 Function Underlies the Functional Abnormalities of MeCP2 Mutant Neurons. Mol Cell 2020; 79:84-98.e9. [PMID: 32526163 PMCID: PMC7375197 DOI: 10.1016/j.molcel.2020.05.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 03/04/2020] [Accepted: 05/12/2020] [Indexed: 12/22/2022]
Abstract
Rett syndrome (RTT), mainly caused by mutations in methyl-CpG binding protein 2 (MeCP2), is one of the most prevalent intellectual disorders without effective therapies. Here, we used 2D and 3D human brain cultures to investigate MeCP2 function. We found that MeCP2 mutations cause severe abnormalities in human interneurons (INs). Surprisingly, treatment with a BET inhibitor, JQ1, rescued the molecular and functional phenotypes of MeCP2 mutant INs. We uncovered that abnormal increases in chromatin binding of BRD4 and enhancer-promoter interactions underlie the abnormal transcription in MeCP2 mutant INs, which were recovered to normal levels by JQ1. We revealed cell-type-specific transcriptome impairment in MeCP2 mutant region-specific human brain organoids that were rescued by JQ1. Finally, JQ1 ameliorated RTT-like phenotypes in mice. These data demonstrate that BRD4 dysregulation is a critical driver for RTT etiology and suggest that targeting BRD4 could be a potential therapeutic opportunity for RTT.
Collapse
Affiliation(s)
- Yangfei Xiang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Benjamin Patterson
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sung-Min Hwang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Eriona Hysolli
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Bilal Cakir
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Wanshan Wang
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Young-Jin Kang
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ethan M Clement
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Mei Zhong
- Department of Cell Biology, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Sang-Hun Lee
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yee Sook Cho
- Regenerative Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 305-806, Republic of Korea
| | - Prabir Patra
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, University of Bridgeport, Bridgeport, CT 06604, USA
| | - Gareth J Sullivan
- Department of Molecular Medicine, Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, Oslo University Hospital and University of Oslo, Oslo 0424, Norway; Department of Pediatric Research, Oslo University Hospital Rikshospitalet, Oslo 0372, Norway
| | - Sherman M Weissman
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
226
|
Sun W, Zhao F, Xu Y, Huang K, Guo X, Zheng B, Liu X, Luo Z, Kong Y, Xu M, Schadendorf D, Chen Y. Chondroitin polymerizing factor (CHPF) promotes development of malignant melanoma through regulation of CDK1. Cell Death Dis 2020; 11:496. [PMID: 32612115 PMCID: PMC7329816 DOI: 10.1038/s41419-020-2526-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 11/24/2022]
Abstract
Chondroitin polymerizing factor (CHPF) is an important member of glycosyltransferases involved in the biosynthesis of chondroitin sulfate (CS). However, the relationship between CHPF and malignant melanoma (MM) is still unknown. In this study, it was demonstrated that CHPF was up-regulated in MM tissues compared with the adjacent normal skin tissues and its high expression was correlated with more advanced T stage. Further investigations indicated that the over-expression/knockdown of CHPF could promote/inhibit proliferation, colony formation and migration of MM cells, while inhibiting/promoting cell apoptosis. Moreover, knockdown of CHPF could also suppress tumorigenicity of MM cells in vivo. RNA-sequencing followed by Ingenuity pathway analysis (IPA) was performed for exploring downstream of CHPF and identified CDK1 as the potential target. Furthermore, our study revealed that knockdown of CDK1 could inhibit development of MM in vitro, and alleviate the CHPF over-expression induced promotion of MM. In conclusion, our study showed, as the first time, CHPF as a tumor promotor for MM, whose function was carried out probably through the regulation of CDK1.
Collapse
Affiliation(s)
- Wei Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany
| | - Yu Xu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Kai Huang
- Brandon Reginal Hospital, HCA Healthcare/USF Morsani College of Medicine, Brandon, FL, USA
| | - Xianling Guo
- Department of Oncology, Dermatology Hospital, Tongji University, Shanghai, China
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
- Tongji University Cancer Center, Shanghai, 200072, PR, China
| | - Biqiang Zheng
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, shanghai, 200032, China
| | - Xin Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Zhiguo Luo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yunyi Kong
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45122, Essen, Germany.
| | - Yong Chen
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.
| |
Collapse
|
227
|
Kalous J, Jansová D, Šušor A. Role of Cyclin-Dependent Kinase 1 in Translational Regulation in the M-Phase. Cells 2020; 9:cells9071568. [PMID: 32605021 PMCID: PMC7408968 DOI: 10.3390/cells9071568] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Cyclin dependent kinase 1 (CDK1) has been primarily identified as a key cell cycle regulator in both mitosis and meiosis. Recently, an extramitotic function of CDK1 emerged when evidence was found that CDK1 is involved in many cellular events that are essential for cell proliferation and survival. In this review we summarize the involvement of CDK1 in the initiation and elongation steps of protein synthesis in the cell. During its activation, CDK1 influences the initiation of protein synthesis, promotes the activity of specific translational initiation factors and affects the functioning of a subset of elongation factors. Our review provides insights into gene expression regulation during the transcriptionally silent M-phase and describes quantitative and qualitative translational changes based on the extramitotic role of the cell cycle master regulator CDK1 to optimize temporal synthesis of proteins to sustain the division-related processes: mitosis and cytokinesis.
Collapse
|
228
|
Leukocyte Nucleolus and Anisakis pegreffii-When Falling Apart Means Falling in Place. Genes (Basel) 2020; 11:genes11060688. [PMID: 32585969 PMCID: PMC7348785 DOI: 10.3390/genes11060688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 11/16/2022] Open
Abstract
The view of the nucleolus as a mere ribosomal factory has been recently expanded, highlighting its essential role in immune and stress-related signalling and orchestrating. It has been shown that the nucleolus structure, formed around nucleolus organiser regions (NORs) and attributed Cajal bodies, is prone to disassembly and reassembly correlated to various physiological and pathological stimuli. To evaluate the effect of parasite stimulus on the structure of the leukocyte nucleolus, we exposed rat peripheral blood mononuclear cells (PBMC) to the crude extract of the nematode A. pegreffii (Anisakidae), and compared the observed changes to the effect of control (RPMI-1640 media), immunosuppressive (MPA) and immunostimulant treatment (bacterial lipopolysaccharide (LPS) and viral analogue polyinosinic:polycytidylic acid (poly I:C)) by confocal microscopy. Poly I:C triggered the most accentuated changes such as nucleolar fragmentation and structural unravelling, LPS induced nucleolus thickening reminiscent of cell activation, while MPA induced disassembly of dense fibrillar and granular components. A. pegreffii crude extract triggered nucleolar segregation, expectedly more enhanced in treatment with a higher dose. This is the first evidence that leukocyte nucleoli already undergo structural changes 12 h post-parasitic stimuli, although these are likely to subside after successful cell activation.
Collapse
|
229
|
Viloria K, Nasteska D, Briant LJB, Heising S, Larner DP, Fine NHF, Ashford FB, da Silva Xavier G, Ramos MJ, Hasib A, Cuozzo F, Manning Fox JE, MacDonald PE, Akerman I, Lavery GG, Flaxman C, Morgan NG, Richardson SJ, Hewison M, Hodson DJ. Vitamin-D-Binding Protein Contributes to the Maintenance of α Cell Function and Glucagon Secretion. Cell Rep 2020; 31:107761. [PMID: 32553153 PMCID: PMC7302426 DOI: 10.1016/j.celrep.2020.107761] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/22/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Vitamin-D-binding protein (DBP) or group-specific component of serum (GC-globulin) carries vitamin D metabolites from the circulation to target tissues. DBP is highly localized to the liver and pancreatic α cells. Although DBP serum levels, gene polymorphisms, and autoantigens have all been associated with diabetes risk, the underlying mechanisms remain unknown. Here, we show that DBP regulates α cell morphology, α cell function, and glucagon secretion. Deletion of DBP leads to smaller and hyperplastic α cells, altered Na+ channel conductance, impaired α cell activation by low glucose, and reduced rates of glucagon secretion both in vivo and in vitro. Mechanistically, this involves reversible changes in islet microfilament abundance and density, as well as changes in glucagon granule distribution. Defects are also seen in β cell and δ cell function. Immunostaining of human pancreata reveals generalized loss of DBP expression as a feature of late-onset and long-standing, but not early-onset, type 1 diabetes. Thus, DBP regulates α cell phenotype, with implications for diabetes pathogenesis.
Collapse
Affiliation(s)
- Katrina Viloria
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Linford J B Briant
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Dean P Larner
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Nicholas H F Fine
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Fiona B Ashford
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Gabriela da Silva Xavier
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Maria Jiménez Ramos
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Annie Hasib
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Federica Cuozzo
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK
| | - Jocelyn E Manning Fox
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Patrick E MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Ildem Akerman
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK
| | - Christine Flaxman
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK
| | - Noel G Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK
| | - Sarah J Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK
| | - Martin Hewison
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK.
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham B15 2TT, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TT, UK; Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
230
|
Hégarat N, Crncec A, Suarez Peredo Rodriguez MF, Echegaray Iturra F, Gu Y, Busby O, Lang PF, Barr AR, Bakal C, Kanemaki MT, Lamond AI, Novak B, Ly T, Hochegger H. Cyclin A triggers Mitosis either via the Greatwall kinase pathway or Cyclin B. EMBO J 2020; 39:e104419. [PMID: 32350921 PMCID: PMC7265243 DOI: 10.15252/embj.2020104419] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 01/23/2023] Open
Abstract
Two mitotic cyclin types, cyclin A and B, exist in higher eukaryotes, but their specialised functions in mitosis are incompletely understood. Using degron tags for rapid inducible protein removal, we analyse how acute depletion of these proteins affects mitosis. Loss of cyclin A in G2-phase prevents mitotic entry. Cells lacking cyclin B can enter mitosis and phosphorylate most mitotic proteins, because of parallel PP2A:B55 phosphatase inactivation by Greatwall kinase. The final barrier to mitotic establishment corresponds to nuclear envelope breakdown, which requires a decisive shift in the balance of cyclin-dependent kinase Cdk1 and PP2A:B55 activity. Beyond this point, cyclin B/Cdk1 is essential for phosphorylation of a distinct subset of mitotic Cdk1 substrates that are essential to complete cell division. Our results identify how cyclin A, cyclin B and Greatwall kinase coordinate mitotic progression by increasing levels of Cdk1-dependent substrate phosphorylation.
Collapse
Affiliation(s)
- Nadia Hégarat
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Adrijana Crncec
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | | | | | - Yan Gu
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Oliver Busby
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| | - Paul F Lang
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Alexis R Barr
- MRC London Institute of Medical ScienceImperial CollegeLondonUK
- Institute of Clinical SciencesFaculty of MedicineImperial CollegeLondonUK
| | - Chris Bakal
- Institute for Cancer ResearchChester Beatty LaboratoriesLondonUK
| | - Masato T Kanemaki
- National Institute of GeneticsResearch Organization of Information and Systems (ROIS)MishimaJapan
- Department of GeneticsSOKENDAI (The Graduate University of Advanced Studies)MishimaJapan
| | - Angus I Lamond
- Centre for Gene Regulation and ExpressionSchool of Life SciencesUniversity of DundeeDundeeUK
| | - Bela Novak
- Department of BiochemistryUniversity of OxfordOxfordUK
| | - Tony Ly
- Wellcome Trust Centre for Cell BiologyUniversity of EdinburghEdinburghUK
| | - Helfrid Hochegger
- Genome Damage and Stability CentreSchool of Life SciencesUniversity of SussexBrightonUK
| |
Collapse
|
231
|
Jackman M, Marcozzi C, Barbiero M, Pardo M, Yu L, Tyson AL, Choudhary JS, Pines J. Cyclin B1-Cdk1 facilitates MAD1 release from the nuclear pore to ensure a robust spindle checkpoint. J Cell Biol 2020; 219:e201907082. [PMID: 32236513 PMCID: PMC7265330 DOI: 10.1083/jcb.201907082] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/05/2020] [Accepted: 03/06/2020] [Indexed: 11/22/2022] Open
Abstract
How the cell rapidly and completely reorganizes its architecture when it divides is a problem that has fascinated researchers for almost 150 yr. We now know that the core regulatory machinery is highly conserved in eukaryotes, but how these multiple protein kinases, protein phosphatases, and ubiquitin ligases are coordinated in space and time to remodel the cell in a matter of minutes remains a major question. Cyclin B1-Cdk is the primary kinase that drives mitotic remodeling; here we show that it is targeted to the nuclear pore complex (NPC) by binding an acidic face of the kinetochore checkpoint protein, MAD1, where it coordinates NPC disassembly with kinetochore assembly. Localized cyclin B1-Cdk1 is needed for the proper release of MAD1 from the embrace of TPR at the nuclear pore so that it can be recruited to kinetochores before nuclear envelope breakdown to maintain genomic stability.
Collapse
|
232
|
Kafer GR, Cesare AJ. A Survey of Essential Genome Stability Genes Reveals That Replication Stress Mitigation Is Critical for Peri-Implantation Embryogenesis. Front Cell Dev Biol 2020; 8:416. [PMID: 32548123 PMCID: PMC7274024 DOI: 10.3389/fcell.2020.00416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
Abstract
Murine development demands that pluripotent epiblast stem cells in the peri-implantation embryo increase from approximately 120 to 14,000 cells between embryonic days (E) 4.5 and E7.5. This is possible because epiblast stem cells can complete cell cycles in under 3 h in vivo. To ensure conceptus fitness, epiblast cells must undertake this proliferative feat while maintaining genome integrity. How epiblast cells maintain genome health under such an immense proliferation demand remains unclear. To illuminate the contribution of genome stability pathways to early mammalian development we systematically reviewed knockout mouse data from 347 DDR and repair associated genes. Cumulatively, the data indicate that while many DNA repair functions are dispensable in embryogenesis, genes encoding replication stress response and homology directed repair factors are essential specifically during the peri-implantation stage of early development. We discuss the significance of these findings in the context of the unique proliferative demands placed on pluripotent epiblast stem cells.
Collapse
Affiliation(s)
| | - Anthony J. Cesare
- Genome Integrity Unit, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
233
|
Seminal Plasma Induces Overexpression of Genes Associated with Embryo Development and Implantation in Day-6 Porcine Blastocysts. Int J Mol Sci 2020; 21:ijms21103662. [PMID: 32455957 PMCID: PMC7279338 DOI: 10.3390/ijms21103662] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 02/08/2023] Open
Abstract
The infusion of boar seminal plasma (SP) before artificial insemination (AI) positively alters the expression of endometrial genes and pathways involved in embryo development. This study aimed to determine which transcriptome changes occur in preimplantation embryos in response to SP infusions during estrus. Postweaning estrus sows received 40-mL intrauterine infusions of either SP (N = 6) or BTS extender (control group; N = 6) 30 min before each of two post-cervical AIs. On Day 6, embryos were surgically collected and analyzed for differential gene expression. Microarray analysis of embryos revealed 210 annotated genes, differentially expressed (p-value < 0.05 and fold change </> 2) in SP-blastocysts, compared to controls. Most of these genes were associated with biological, cellular, metabolic and developmental processes. The pathways enriched among the upregulated genes related to signal transduction, cellular processes and the endocrine system. Among altered genes involved in these pathways, the SP-group showed a conspicuous overexpression of ApoA-I, CDK1, MAPK1, SMAD2, PRKAA1 and RICTOR, with reported key roles in embryo development, implantation, or progression of pregnancy. In conclusion, the results demonstrate that SP infusions prior to AI upregulates the expression of embryo development related genes in Day 6 pig embryos.
Collapse
|
234
|
Levada K, Pshenichnikov S, Omelyanchik A, Rodionova V, Nikitin A, Savchenko A, Schetinin I, Zhukov D, Abakumov M, Majouga A, Lunova M, Jirsa M, Smolková B, Uzhytchak M, Dejneka A, Lunov O. Progressive lysosomal membrane permeabilization induced by iron oxide nanoparticles drives hepatic cell autophagy and apoptosis. NANO CONVERGENCE 2020; 7:17. [PMID: 32424769 PMCID: PMC7235155 DOI: 10.1186/s40580-020-00228-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Iron oxide nanoparticles (IONs) are frequently used in various biomedical applications, in particular as magnetic resonance imaging contrast agents in liver imaging. Indeed, number of IONs have been withdrawn due to their poor clinical performance. Yet comprehensive understanding of their interactions with hepatocytes remains relatively limited. Here we investigated how iron oxide nanocubes (IO-cubes) and clusters of nanocubes (IO-clusters) affect distinct human hepatic cell lines. The viability of HepG2, Huh7 and Alexander cells was concentration-dependently decreased after exposure to either IO-cubes or IO-clusters. We found similar cytotoxicity levels in three cell lines triggered by both nanoparticle formulations. Our data indicate that different expression levels of Bcl-2 predispose cell death signaling mediated by nanoparticles. Both nanoparticles induced rather apoptosis than autophagy in HepG2. Contrary, IO-cubes and IO-clusters trigger distinct cell death signaling events in Alexander and Huh7 cells. Our data clarifies the mechanism by which cubic nanoparticles induce autophagic flux and the mechanism of subsequent toxicity. These findings imply that the cytotoxicity of ION-based contrast agents should be carefully considered, particularly in patients with liver diseases.
Collapse
Affiliation(s)
- Kateryna Levada
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Stanislav Pshenichnikov
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Alexander Omelyanchik
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Valeria Rodionova
- Institute of Physics, Mathematics and Information Technology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Aleksey Nikitin
- National University of Science and Technology "MISIS", Moscow, Russia
| | | | - Igor Schetinin
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Dmitry Zhukov
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Maxim Abakumov
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Alexander Majouga
- National University of Science and Technology "MISIS", Moscow, Russia
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221, Prague, Czech Republic.
| |
Collapse
|
235
|
The crucial p53-dependent oncogenic role of JAB1 in osteosarcoma in vivo. Oncogene 2020; 39:4581-4591. [PMID: 32390003 PMCID: PMC7274902 DOI: 10.1038/s41388-020-1320-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 12/25/2022]
Abstract
Osteosarcoma (OS) is the most common primary bone cancer and ranks amongst the leading causes of cancer mortality in young adults. Jun activation domain binding protein 1 (JAB1) is overexpressed in many cancers and has recently emerged as a novel target for cancer treatment. However, the role of JAB1 in osteosarcoma was virtually unknown. In this study, we demonstrate that JAB1-knockdown in malignant osteosarcoma cell lines significantly reduced their oncogenic properties, including proliferation, colony formation, and motility. We also performed RNA-sequencing analysis in JAB1-knockdown OS cells and identified 4110 genes that are significantly differentially expressed. This demonstrated for the first time that JAB1 regulates a large and specific transcriptome in cancer. We also found that JAB1 is overexpressed in human OS and correlates with a poor prognosis. Moreover, we generated a novel mouse model that overexpresses Jab1 specifically in osteoblasts upon a TP53 heterozygous sensitizing background. Interestingly, by 13 months of age, a significant proportion of these mice spontaneously developed conventional OS. Finally, we demonstrate that a novel, highly specific small molecule inhibitor of JAB1, CSN5i-3, reduces osteosarcoma cell viability and has specific effects on the ubiquitin-proteasome system in OS. Thus, we show for the first time that the overexpression of JAB1 in vivo can result in accelerated spontaneous tumor formation in a p53-dependent manner. In summary, JAB1 might be a unique target for the treatment of osteosarcoma and other cancers.
Collapse
|
236
|
Huang XM, Yang WC, Liu Y, Tang DR, Wu T, Sun FY. Mutations in MC4R facilitate the angiogenic activity in patients with orbital venous malformation. Exp Biol Med (Maywood) 2020; 245:956-963. [PMID: 32363922 DOI: 10.1177/1535370220919056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPACT STATEMENT The detailed molecular mechanism of orbital venous malformation (OVM) is still not clear. Using whole exome sequencing, 4 types of melanocortin 4 receptor (MC4R) mutation were detected in 7 of 27 patients with OVM, and all types of MC4R mutations resulted in the upregulation of MC4R expression. In vitro study indicated that MC4R has impacts on the proliferation, cell cycle, migration, and tube formation of the endothelial cells. Moreover, MC4R mutations altered the downstream signaling, including cAMP concentration and the expression levels of several PI3K/AKT/mTOR downstream genes, including p21, cyclin B1, ITGA10, and ITGA11. MC4R mutations may lead to the pathogenesis of OVM through modulating the downstream signaling to alter the angiogenic activity of endothelial cells.
Collapse
Affiliation(s)
- Xiao-Ming Huang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China.,Orbital Disease Institute, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Wan-Chen Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Yang Liu
- Orbital Disease Institute, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Dong-Run Tang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Tong Wu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Feng-Yuan Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| |
Collapse
|
237
|
Michowski W, Chick JM, Chu C, Kolodziejczyk A, Wang Y, Suski JM, Abraham B, Anders L, Day D, Dunkl LM, Li Cheong Man M, Zhang T, Laphanuwat P, Bacon NA, Liu L, Fassl A, Sharma S, Otto T, Jecrois E, Han R, Sweeney KE, Marro S, Wernig M, Geng Y, Moses A, Li C, Gygi SP, Young RA, Sicinski P. Cdk1 Controls Global Epigenetic Landscape in Embryonic Stem Cells. Mol Cell 2020; 78:459-476.e13. [PMID: 32240602 DOI: 10.1016/j.molcel.2020.03.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/26/2020] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
The cyclin-dependent kinase 1 (Cdk1) drives cell division. To uncover additional functions of Cdk1, we generated knockin mice expressing an analog-sensitive version of Cdk1 in place of wild-type Cdk1. In our study, we focused on embryonic stem cells (ESCs), because this cell type displays particularly high Cdk1 activity. We found that in ESCs, a large fraction of Cdk1 substrates is localized on chromatin. Cdk1 phosphorylates many proteins involved in epigenetic regulation, including writers and erasers of all major histone marks. Consistent with these findings, inhibition of Cdk1 altered histone-modification status of ESCs. High levels of Cdk1 in ESCs phosphorylate and partially inactivate Dot1l, the H3K79 methyltransferase responsible for placing activating marks on gene bodies. Decrease of Cdk1 activity during ESC differentiation de-represses Dot1l, thereby allowing coordinated expression of differentiation genes. These analyses indicate that Cdk1 functions to maintain the epigenetic identity of ESCs.
Collapse
Affiliation(s)
- Wojciech Michowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Joel M Chick
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Chen Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aleksandra Kolodziejczyk
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Yichen Wang
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Jan M Suski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brian Abraham
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lars Anders
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel Day
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Lukas M Dunkl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mitchell Li Cheong Man
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada
| | - Tian Zhang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Phatthamon Laphanuwat
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nickolas A Bacon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lijun Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Samanta Sharma
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Tobias Otto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Emanuelle Jecrois
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Richard Han
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Katharine E Sweeney
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Samuele Marro
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine and Department of Pathology, Stanford University Medical School, Stanford, CA 94305, USA
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alan Moses
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; Center for Analysis of Genome Evolution and Function, Toronto, ON M5S 3B2, Canada
| | - Cheng Li
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
238
|
Identification of cancer stem cell-related biomarkers in lung adenocarcinoma by stemness index and weighted correlation network analysis. J Cancer Res Clin Oncol 2020; 146:1463-1472. [PMID: 32221746 DOI: 10.1007/s00432-020-03194-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Accounting for tumor heterogeneity, cancer stem cells (CSC) are involved in tumor metastasis, relapse, and drug resistance. Genes regulating CSC characteristics in lung adenocarcinoma (LUAD) were explored and validated in this study. METHODS The mRNA stemness index (mRNAsi) of more than 500 LUAD cases from The Cancer Genome Atlas database were calculated using a one-class logistic regression machine learning algorithm based on the mRNA expression of pluripotent stem cells and their differentiated progeny. mRNAsi-related key genes were identified by weighted correlation network analysis. The expression levels and prognostic roles of key genes were analyzed in Oncomine, PrognoScan, and Kaplan-Meier plotter databases, and validated using data from our center. RESULTS The mRNAsi was significantly higher in LUAD compared with normal lung tissues. LUAD patients of advanced stage exhibited a higher mRNAsi and worse overall survival (OS). Eight key genes were identified: heat shock 70 kDa protein 4 (HSPA4), cell division cycle associated 7 (CDCA7), cell division cycle 20 (CDC20), cyclin-dependent kinase 1 (CDK1), CAP-GLY domain containing linker protein 1 (CLIP1), cyclin B1 (CCNB1), H2A histone family, member X (H2AFX), and Bloom syndrome, RecQ helicase-like (BLM). These genes were differentially expressed in various types of malignancies and validated in the LUAD cases. LUAD patients with low expression of CDC20, CDK1, CCNB1, H2AFX, or BLM had a significantly better OS, whereas OS was reduced for patients with low expression of CLIP1. In addition, the expression of CDCA7 did not significantly impact the OS of LUAD patients. The protein-protein interaction networks evaluated by STRING demonstrated strong relationships between these key genes, which were validated in our cases. CONCLUSIONS The mRNAsi was significantly higher in LUAD compared with normal samples. Eight mRNAsi-related key genes were associated with prognosis and the cell cycle, and were strongly correlated with each other and differentially expressed in tumor and normal samples. We provide a new strategy for exploring stemness-related genes in LUAD cases.
Collapse
|
239
|
Ding L, Cao J, Lin W, Chen H, Xiong X, Ao H, Yu M, Lin J, Cui Q. The Roles of Cyclin-Dependent Kinases in Cell-Cycle Progression and Therapeutic Strategies in Human Breast Cancer. Int J Mol Sci 2020; 21:ijms21061960. [PMID: 32183020 PMCID: PMC7139603 DOI: 10.3390/ijms21061960] [Citation(s) in RCA: 365] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) are serine/threonine kinases whose catalytic activities are regulated by interactions with cyclins and CDK inhibitors (CKIs). CDKs are key regulatory enzymes involved in cell proliferation through regulating cell-cycle checkpoints and transcriptional events in response to extracellular and intracellular signals. Not surprisingly, the dysregulation of CDKs is a hallmark of cancers, and inhibition of specific members is considered an attractive target in cancer therapy. In breast cancer (BC), dual CDK4/6 inhibitors, palbociclib, ribociclib, and abemaciclib, combined with other agents, were approved by the Food and Drug Administration (FDA) recently for the treatment of hormone receptor positive (HR+) advanced or metastatic breast cancer (A/MBC), as well as other sub-types of breast cancer. Furthermore, ongoing studies identified more selective CDK inhibitors as promising clinical targets. In this review, we focus on the roles of CDKs in driving cell-cycle progression, cell-cycle checkpoints, and transcriptional regulation, a highlight of dysregulated CDK activation in BC. We also discuss the most relevant CDK inhibitors currently in clinical BC trials, with special emphasis on CDK4/6 inhibitors used for the treatment of estrogen receptor-positive (ER+)/human epidermal growth factor 2-negative (HER2−) M/ABC patients, as well as more emerging precise therapeutic strategies, such as combination therapies and microRNA (miRNA) therapy.
Collapse
Affiliation(s)
- Lei Ding
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Jiaqi Cao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Wen Lin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Hongjian Chen
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Xianhui Xiong
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Hongshun Ao
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Min Yu
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Jie Lin
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
| | - Qinghua Cui
- Lab of Biochemistry & Molecular Biology, School of Life Sciences, Yunnan University, Kunming 650091, China; (L.D.); (J.C.); (W.L.); (H.C.); (X.X.); (H.A.); (M.Y.); (J.L.)
- Key Lab of Molecular Cancer Biology, Yunnan Education Department, Kunming 650091, China
- Correspondence:
| |
Collapse
|
240
|
Ma Z, Yang J, Yang Y, Wang X, Chen G, Shi A, Lu Y, Jia S, Kang X, Lu L. Rosmarinic acid exerts an anticancer effect on osteosarcoma cells by inhibiting DJ-1 via regulation of the PTEN-PI3K-Akt signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153186. [PMID: 32088353 DOI: 10.1016/j.phymed.2020.153186] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/02/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Osteosarcoma is the most common type of primary malignant bone tumor. This disease has exhibited a progressively lower survival rate over the past several decades, which has resulted in it becoming a main cause of death in humans. Rosmarinic acid (RA), a water-soluble polyphenolic phytochemical, exerts powerful anticancer effects against multiple types of cancer; however, its potential effects on osteosarcoma remain unknown. Hence, the present study investigated the efficacy of RA against osteosarcoma and aimed to clarify the mechanisms underlying this process. METHODS The effects of RA on cell viability, apoptosis, cell cycle distribution, migration, invasion, and signaling molecules were analyzed by CCK-8 assay, flowcytometric analysis, wound healing assay, Transwell assay, proteomic analysis, and use of shRNAs. RESULTS RA exerted anti-proliferation and pro-apoptotic effects on U2OS and MG63 osteosarcoma cells. Apoptosis was induced via extrinsic and intrinsic pathways by increasing the Bax/Bcl-2 ratio, triggering the intracellular production of reactive oxygen species (ROS), reducing the mitochondrial membrane potential (MMP), and upregulating the cleavage rates of caspase-8, caspase-9, and caspase-3. Additionally, RA suppressed the migration and invasion of osteosarcoma cells by inhibiting the expression levels of matrix metalloproteinase-2 and -9 (MMP-2 and -9), which are associated with a weakening of the epithelial-mesenchymal transition (EMT). Moreover, proteomic analyses identified DJ-1 as a potential target for RA. Several studies have indicated an oncogenic role for DJ-1 using knockdowns via the lentiviral-mediated transfection of shRNA, which caused the conspicuous suppression of cell proliferation, migration, and invasion as well as the arrest of cell cycle progression. At the molecular level, the expression levels of DJ-1, p-PI3K, and p-Akt were reduced, whereas the protein levels of phosphatase and tensin homologue (PTEN) were increased. CONCLUSION In conjunction with the high levels of DJ-1 expression in osteosarcoma tissues and cell lines, the present results suggested that RA exhibited anticancer effects in osteosarcoma cells by inhibiting DJ-1 via regulation of the PTEN-PI3K-Akt signaling pathway. Therefore, DJ-1 might be a biological target for RA in osteosarcoma cells.
Collapse
Affiliation(s)
- Zhanjun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, China
| | - Jingjing Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yang Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Xuexi Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, Gansu 730000, China; School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Guohu Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ancheng Shi
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yubao Lu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Shouning Jia
- Traditional Chinese Medicine Hospital of Qinghai Province, Xining, Qinghai 810000, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, China.
| | - Li Lu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou, Gansu 730000, China; Institute of Pharmacology, School of Basic Medical Science, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
241
|
Basu S, Roberts EL, Jones AW, Swaffer MP, Snijders AP, Nurse P. The Hydrophobic Patch Directs Cyclin B to Centrosomes to Promote Global CDK Phosphorylation at Mitosis. Curr Biol 2020; 30:883-892.e4. [PMID: 32084401 PMCID: PMC7063568 DOI: 10.1016/j.cub.2019.12.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/20/2019] [Accepted: 12/17/2019] [Indexed: 11/15/2022]
Abstract
The cyclin-dependent kinases (CDKs) are the major cell-cycle regulators that phosphorylate hundreds of substrates, controlling the onset of S phase and M phase [1, 2, 3]. However, the patterns of substrate phosphorylation increase are not uniform, as different substrates become phosphorylated at different times as cells proceed through the cell cycle [4, 5]. In fission yeast, the correct ordering of CDK substrate phosphorylation can be established by the activity of a single mitotic cyclin-CDK complex [6, 7]. Here, we investigate the substrate-docking region, the hydrophobic patch, on the fission yeast mitotic cyclin Cdc13 as a potential mechanism to correctly order CDK substrate phosphorylation. We show that the hydrophobic patch targets Cdc13 to the yeast centrosome equivalent, the spindle pole body (SPB), and disruption of this motif prevents both centrosomal localization of Cdc13 and the onset of mitosis but does not prevent S phase. CDK phosphorylation in mitosis is compromised for approximately half of all mitotic CDK substrates, with substrates affected generally being those that require the highest levels of CDK activity to become phosphorylated and those that are located at the SPB. Our experiments suggest that the hydrophobic patch of mitotic cyclins contributes to CDK substrate selection by directing the localization of Cdc13-CDK to centrosomes and that this localization of CDK contributes to the CDK substrate phosphorylation necessary to ensure proper entry into mitosis. Finally, we show that mutation of the hydrophobic patch prevents cyclin B1 localization to centrosomes in human cells, suggesting that this mechanism of cyclin-CDK spatial regulation may be conserved across eukaryotes. The hydrophobic patch of human and yeast cyclin B directs it to the centrosome Loss of the yeast cyclin B hydrophobic patch allows S phase but prevents mitosis Compartmentalized mitotic CDK phosphorylation relies on the hydrophobic patch
Collapse
Affiliation(s)
- Souradeep Basu
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| | - Emma L Roberts
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Andrew W Jones
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Protein Analysis and Proteomics Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Matthew P Swaffer
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Ambrosius P Snijders
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London NW1 1AT, UK
| | - Paul Nurse
- Cell Cycle Laboratory, The Francis Crick Institute, London NW1 1AT, UK; Laboratory of Yeast Genetics and Cell Biology, Rockefeller University, New York, NY 10065, USA
| |
Collapse
|
242
|
Ijomone OM, Miah MR, Akingbade GT, Bucinca H, Aschner M. Nickel-Induced Developmental Neurotoxicity in C. elegans Includes Cholinergic, Dopaminergic and GABAergic Degeneration, Altered Behaviour, and Increased SKN-1 Activity. Neurotox Res 2020; 37:1018-1028. [DOI: 10.1007/s12640-020-00175-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/13/2020] [Accepted: 02/02/2020] [Indexed: 12/11/2022]
|
243
|
Luo Y, Wang L, Ran W, Li G, Xiao Y, Wang X, Zhao H, Xing X. Overexpression of SAPCD2 correlates with proliferation and invasion of colorectal carcinoma cells. Cancer Cell Int 2020; 20:43. [PMID: 32055236 PMCID: PMC7006392 DOI: 10.1186/s12935-020-1121-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Background Suppressor anaphase-promoting complex domain containing 2 (SAPCD2) is a novel gene playing important roles in the initiation, invasion, and metastasis of several malignancies. However, its role in colorectal carcinoma (CRC) still remains unclear. Method In this study, we investigated the expression and biological function of SAPCD2 in CRC. Immunohistochemistry (IHC) for SAPCD2 was performed in 410 pairs of CRC specimens and corresponding normal epithelial tissues, and in 50 adenoma tissues. Clinical pathological factors were analyzed in relation to the expression of SAPCD2. The biological functions of SAPCD2 in CRC cells and its effect on cell cycle were investigated in vitro and in vivo through gain/loss-of-function approaches. Results IHC showed that SAPCD2 expression was significantly higher in CRC tissues compared to adenoma and normal epithelium tissues and was correlated with tumor location (p = 0.018). SAPCD2 significantly promoted cell proliferation, migration, and invasion both in vitro and in vivo (p < 0.05). In addition, SAPCD2 knockdown in CRC cells was associated with reduced G1/S transition, while overexpression caused G2/M phase arrest (p < 0.05). Conclusions In sum, SAPCD2 is overexpressed in CRC tissues and plays a critical role in CRC progression. Therefore, it might represent a promising therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Yage Luo
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China.,2Department of Pathology, Qingdao University Basic Medicine College, Qingdao, 266000 Shandong People's Republic of China
| | - Lili Wang
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Wenwen Ran
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Guangqi Li
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Yujing Xiao
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Xiaonan Wang
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Han Zhao
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| | - Xiaoming Xing
- 1Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000 Shandong People's Republic of China
| |
Collapse
|
244
|
Lv S, Ning H, Li Y, Wang J, Jia Q, Wen H. Inhibition of cyclinB1 Suppressed the Proliferation, Invasion, and Epithelial Mesenchymal Transition of Hepatocellular Carcinoma Cells and Enhanced the Sensitivity to TRAIL-Induced Apoptosis. Onco Targets Ther 2020; 13:1119-1128. [PMID: 32103981 PMCID: PMC7008233 DOI: 10.2147/ott.s225202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Background CyclinB1 is highly expressed in various tumor tissues and plays an important role in tumor progression. However, its role in hepatocellular carcinoma (HCC) remains unclear. Therefore, the aim of this study was to explore the role of cyclinB1 in the development and progression of HCC. Methods The expression of cyclinB1 was analyzed using the Gene Expression Profiling Interactive Analysis (GEPIA) database, and detected in HCC tissues and HCC cell lines through quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blotting. CyclinB1-short hairpin RNA (Sh-cyclinB1) was transfected into HCC cells to knockdown cyclinB1, and the effect of cyclinB1 knockdown on HCC was examined via the MTT assay, colony formation assay, wound healing assay, scratch assay, cell cycle analysis in vitro, and xenograft model in nude mice. In addition, the role of cyclinB1 on tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis was measured using flow cytometry and Western blotting. Results The GEPIA database analysis showed that cyclinB1 was highly expressed in HCC tissues. The results of qRT-PCR and Western blotting proved that the expression of cyclinB1 was significantly increased in HCC tissues and cell lines. The data of the MTT assay, colony formation assay, and cell cycle analysis indicated that cyclinB1 knockdown inhibited the proliferation of HCC cells. In addition, cell migration, invasion, and epithelial mesenchymal transition were also impaired by cyclinB1 knockdown. Furthermore, the xenograft model in nude mice demonstrated that inhibition of cyclinB1 suppressed tumor growth and metastasis in vivo. Finally, the results of flow cytometry and Western blotting indicated that inhibition of cyclinB1 enhanced the sensitivity of HCC cells to TRAIL-induced apoptosis. Conclusion Overall, these data provide reasonable evidence that cyclinB1 may serve as a proto-oncogene during the progression of HCC.
Collapse
Affiliation(s)
- Shuai Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| | - Hanbing Ning
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| | - Yingxia Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| | - Jingyun Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| | - Qiaoyu Jia
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| | - Hongtao Wen
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450018, Henan Province, People's Republic of China
| |
Collapse
|
245
|
Kim HM, He L, Lee S, Park C, Kim DH, Han HJ, Han J, Hwang J, Cha-Molstad H, Lee KH, Ko SK, Jang JH, Ryoo IJ, Blenis J, Lee HG, Ahn JS, Kwon YT, Soung NK, Kim BY. Inhibition of osteoclasts differentiation by CDC2-induced NFATc1 phosphorylation. Bone 2020; 131:115153. [PMID: 31730830 DOI: 10.1016/j.bone.2019.115153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/18/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Bone homeostasis is regulated by a balance of bone formation and bone resorption; dysregulation of bone homeostasis may cause bone-related diseases (e.g., osteoporosis, osteopetrosis, bone fracture). Members of the nuclear factor of activated T cells (NFAT) family of transcription factors play crucial roles in the regulation of immune system, inflammatory responses, cardiac formation, skeletal muscle development, and bone homeostasis. Of these, NFATc1 is a key transcription factor mediating osteoclast differentiation, which is regulated by phosphorylation by distinct NFAT kinases including casein kinase 1 (CK1), glycogen synthase kinase 3 (GSK3), and dual-specificity tyrosine-phosphorylation-regulated kinases (DYRKs). In this study, we report that cell division control protein 2 homolog (cdc2) is a novel NFAT protein kinase that inhibits NFATc1 activation by direct phosphorylation of the NFATc1 S263 residue. Cdc2 inhibitors such as Roscovitine and BMI-1026 induce reduction of phosphorylation of NFATc1, and this process leads to the inhibition of NFATc1 translocation from the nucleus to the cytoplasm, consequently increasing the nuclear pool of NFATc1. Additionally, the inhibition of cdc2-mediated NFATc1 phosphorylation causes an elevation of osteoclast differentiation or TRAP-positive staining in zebrafish scales. Our results suggest that cdc2 is a novel NFAT protein kinase that negatively regulates osteoclast differentiation.
Collapse
Affiliation(s)
- Hye-Min Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Long He
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sangku Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Chanmi Park
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Dong Hyun Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Ho-Jin Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Junyeol Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Joonsung Hwang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Hyunjoo Cha-Molstad
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Sung-Kyun Ko
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - Jae-Hyuk Jang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - In-Ja Ryoo
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Jong Seog Ahn
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea.
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Bo Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
246
|
Fu R, Guo H, Janga S, Choi M, Klinngam W, Edman MC, Hamm-Alvarez SF. Cathepsin S activation contributes to elevated CX3CL1 (fractalkine) levels in tears of a Sjögren's syndrome murine model. Sci Rep 2020; 10:1455. [PMID: 31996771 PMCID: PMC6989636 DOI: 10.1038/s41598-020-58337-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/14/2020] [Indexed: 11/09/2022] Open
Abstract
Autoimmune dacryoadenitis and altered lacrimal gland (LG) secretion are features of Sjögren's syndrome (SS). Activity of cathepsin S (CTSS), a cysteine protease, is significantly and specifically increased in SS patient tears. The soluble chemokine, CX3CL1 (fractalkine), is cleaved from membrane-bound CX3CL1 by proteases including CTSS. We show that CX3CL1 is significantly elevated by 2.5-fold in tears (p = 0.0116) and 1.4-fold in LG acinar cells (LGAC)(p = 0.0026) from male NOD mice, a model of autoimmune dacryoadenitis in SS, relative to BALB/c controls. Primary mouse LGAC and human corneal epithelial cells (HCE-T cells) exposed to interferon-gamma, a cytokine elevated in SS, showed up to 9.6-fold (p ≤ 0.0001) and 25-fold (p ≤ 0.0001) increases in CX3CL1 gene expression, and 1.9-fold (p = 0.0005) and 196-fold (p ≤ 0.0001) increases in CX3CL1 protein expression, respectively. Moreover, exposure of HCE-T cells to recombinant human CTSS at activity equivalent to that in SS patient tears increased cellular CX3CL1 gene and protein expression by 2.8-fold (p = 0.0021) and 5.1-fold (p ≤ 0.0001), while increasing CX3CL1 in culture medium by 5.8-fold (p ≤ 0.0001). Flow cytometry demonstrated a 4.5-fold increase in CX3CR1-expressing immune cells (p ≤ 0.0001), including increased T-cells and macrophages, in LG from NOD mice relative to BALB/c. CTSS-mediated induction/cleavage of CX3CL1 may contribute to ocular surface and LG inflammation in SS.
Collapse
Affiliation(s)
- Runzhong Fu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Srikanth Janga
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Minchang Choi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Wannita Klinngam
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Maria C Edman
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Sarah F Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
247
|
Vegi NM, Chakrabortty S, Zegota MM, Kuan SL, Stumper A, Rawat VPS, Sieste S, Buske C, Rau S, Weil T, Feuring-Buske M. Somatostatin receptor mediated targeting of acute myeloid leukemia by photodynamic metal complexes for light induced apoptosis. Sci Rep 2020; 10:371. [PMID: 31941913 PMCID: PMC6962389 DOI: 10.1038/s41598-019-57172-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 12/21/2019] [Indexed: 02/08/2023] Open
Abstract
Acute myeloid leukemia (AML) is characterized by relapse and treatment resistance in a major fraction of patients, underlining the need of innovative AML targeting therapies. Here we analysed the therapeutic potential of an innovative biohybrid consisting of the tumor-associated peptide somatostatin and the photosensitizer ruthenium in AML cell lines and primary AML patient samples. Selective toxicity was analyzed by using CD34 enriched cord blood cells as control. Treatment of OCI AML3, HL60 and THP1 resulted in a 92, and 99 and 97% decrease in clonogenic growth compared to the controls. Primary AML cells demonstrated a major response with a 74 to 99% reduction in clonogenicity in 5 of 6 patient samples. In contrast, treatment of CD34+ CB cells resulted in substantially less reduction in colony numbers. Subcellular localization assays of RU-SST in OCI-AML3 cells confirmed strong co-localization of RU-SST in the lysosomes compared to the other cellular organelles. Our data demonstrate that conjugation of a Ruthenium complex with somatostatin is efficiently eradicating LSC candidates of patients with AML. This indicates that receptor mediated lysosomal accumulation of photodynamic metal complexes is a highly attractive approach for targeting AML cells.
Collapse
Affiliation(s)
- Naidu M Vegi
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Sabyasachi Chakrabortty
- Department of Chemistry, SRM University, AP - Amaravati, Andhra Pradesh, 522502, India.,Max Planck Institute for Polymer Research, D-55128, Mainz, Germany
| | - Maksymilian M Zegota
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Anne Stumper
- Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Vijay P S Rawat
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Stefanie Sieste
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Christian Buske
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany
| | - Sven Rau
- Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, D-55128, Mainz, Germany.,Institute of Inorganic Chemistry I, Ulm University, D-89081, Ulm, Germany
| | - Michaela Feuring-Buske
- Institute of Experimental Cancer Research, Comprehensive Cancer Centre, University Hospital Ulm, D-89081, Ulm, Germany. .,Department of Internal Medicine III, University Hospital Ulm, D-89081, Ulm, Germany.
| |
Collapse
|
248
|
Serres MP, Samwer M, Truong Quang BA, Lavoie G, Perera U, Görlich D, Charras G, Petronczki M, Roux PP, Paluch EK. F-Actin Interactome Reveals Vimentin as a Key Regulator of Actin Organization and Cell Mechanics in Mitosis. Dev Cell 2020; 52:210-222.e7. [PMID: 31928973 PMCID: PMC6983945 DOI: 10.1016/j.devcel.2019.12.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 09/23/2019] [Accepted: 12/12/2019] [Indexed: 01/27/2023]
Abstract
Most metazoan cells entering mitosis undergo characteristic rounding, which is important for accurate spindle positioning and chromosome separation. Rounding is driven by contractile tension generated by myosin motors in the sub-membranous actin cortex. Recent studies highlight that alongside myosin activity, cortical actin organization is a key regulator of cortex tension. Yet, how mitotic actin organization is controlled remains poorly understood. To address this, we characterized the F-actin interactome in spread interphase and round mitotic cells. Using super-resolution microscopy, we then screened for regulators of cortex architecture and identified the intermediate filament vimentin and the actin-vimentin linker plectin as unexpected candidates. We found that vimentin is recruited to the mitotic cortex in a plectin-dependent manner. We then showed that cortical vimentin controls actin network organization and mechanics in mitosis and is required for successful cell division in confinement. Together, our study highlights crucial interactions between cytoskeletal networks during cell division. Comparison of the F-actin interactome in spread interphase and round mitotic cells Proteomics identifies vimentin and plectin as key regulators of the mitotic cortex Vimentin intermediate filaments localize under the actin cortex in mitosis Sub-cortical vimentin regulates actin cortex organization and mechanics in mitosis
Collapse
Affiliation(s)
- Murielle P Serres
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK; Cancer Research UK London Research Institute, Clare Hall Laboratories, South Mimms, Hertfordshire, EN6 3LD, UK
| | - Matthias Samwer
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Binh An Truong Quang
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Geneviève Lavoie
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal QC, H3T 1J4, Canada
| | - Upamali Perera
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Guillaume Charras
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK; Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Mark Petronczki
- Cancer Research UK London Research Institute, Clare Hall Laboratories, South Mimms, Hertfordshire, EN6 3LD, UK
| | - Philippe P Roux
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal QC, H3T 1J4, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, QC, H3T 1J4, Canada.
| | - Ewa K Paluch
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
249
|
Day P, Alves N, Daniell E, Dasgupta D, Ogborne R, Steeper A, Raza M, Ellis C, Fawcett J, Keynes R, Muir E. Targeting chondroitinase ABC to axons enhances the ability of chondroitinase to promote neurite outgrowth and sprouting. PLoS One 2020; 15:e0221851. [PMID: 31961897 PMCID: PMC6974052 DOI: 10.1371/journal.pone.0221851] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 08/18/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is currently no effective treatment for promoting regeneration of injured nerves in patients who have sustained injury to the central nervous system such as spinal cord injury. Chondroitinase ABC is an enzyme, which promotes neurite outgrowth and regeneration. It has shown considerable promise as a therapy for these conditions. The aim of the study is to determine if targeting chondroitinase ABC expression to the neuronal axon can further enhance its ability to promote axon outgrowth. Long-distance axon regeneration has not yet been achieved, and would be a significant step in attaining functional recovery following spinal cord injury. METHODOLOGY/PRINCIPAL FINDINGS To investigate this, neuronal cultures were transfected with constructs encoding axon-targeted chondroitinase, non-targeted chondroitinase or GFP, and the effects on neuron outgrowth and sprouting determined on substrates either permissive or inhibitory to neuron regeneration. The mechanisms underlying the observed effects were also explored. Targeting chondroitinase to the neuronal axon markedly enhances its ability to promote neurite outgrowth. The increase in neurite length is associated with an upregulation of β-integrin staining at the axonal cell surface. Staining for phosphofocal adhesion kinase, is also increased, indicating that the β-integrins are in an activated state. Expression of chondroitinase within the neurons also resulted in a decrease in expression of PTEN and RhoA, molecules which present a block to neurite outgrowth, thus identifying two of the pathways by which ChABC promotes neurite outgrowth. CONCLUSIONS / SIGNIFICANCE The novel finding that targeting ChABC to the axon significantly enhances its ability to promote neurite extension, suggests that this may be an effective way of promoting long-distance axon regeneration following spinal cord injury. It could also potentially improve its efficacy in the treatment of other pathologies, where it has been shown to promote recovery, such as myocardial infarction, stroke and Parkinson's disease.
Collapse
Affiliation(s)
- Priscilla Day
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Nuno Alves
- John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Esther Daniell
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Debayan Dasgupta
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rosalie Ogborne
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ashley Steeper
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Mansoor Raza
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Clare Ellis
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - James Fawcett
- John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Roger Keynes
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Elizabeth Muir
- Department of Physiology Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
250
|
Poroca DR, Amer N, Li A, Hanrahan JW, Chappe VM. Changes in the R-region interactions depend on phosphorylation and contribute to PKA and PKC regulation of the cystic fibrosis transmembrane conductance regulator chloride channel. FASEB Bioadv 2020; 2:33-48. [PMID: 32123855 PMCID: PMC6996395 DOI: 10.1096/fba.2019-00053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/19/2019] [Indexed: 01/30/2023] Open
Abstract
The CFTR chloride channel is regulated by phosphorylation at PKA and PKC consensus sites within its regulatory region (R-region) through a mechanism, which is still not completely understood. We used a split-CFTR construct expressing the N-term-TMD1-NBD1 (Front Half; FH), TMD2-NBD2-C-Term (Back Half; BH), and the R-region as separate polypeptides (Split-R) in BHK cells, to investigate in situ how different phosphorylation conditions affect the R-region interactions with other parts of the protein. In proximity ligation assays, we studied the formation of complexes between the R-region and each half of the Split-CFTR. We found that at basal conditions, the density of complexes formed between the R-region and both halves of the split channel were equal. PKC stimulation alone had no effect, whereas PKA stimulation induced the formation of more complexes between the R-region and both halves compared to basal conditions. Moreover, PKC + PKA stimulation further enhanced the formation of FH-R complexes by 40% from PKA level. In cells expressing the Split-R with the two inhibitory PKC sites on the R-region inactivated (SR-S641A/T682A), density of FH-R complexes was much higher than in Split-R WT expressing cells after PKC or PKC + PKA stimulation. No differences were observed for BH-R complexes measured at all phosphorylation conditions. Since full-length CFTR channels display large functional responses to PKC + PKA in WT and S641A/T682A mutant, we conclude that FH-R interactions are important for CFTR function. Inactivation of consensus PKC site serine 686 (S686A) significantly reduced the basal BH-R interaction and prevented the PKC enhancing effect on CFTR function and FH-R interaction. The phospho-mimetic mutation (S686D) restored basal BH-R interaction and the PKC enhancing effect on CFTR function with enhanced FH-R interaction. As the channel function is mainly stimulated by PKA phosphorylation of the R-region, and this response is known to be enhanced by PKC phosphorylation, our data support a model in which the regulation of CFTR activation results from increased interactions of the R-region with the N-term-TMD1-NBD1. Also, serine S686 was found to be critical for the PKC enhancing effect which requires a permissive BH-R interaction at basal level and increased FH-R interaction after PKC + PKA phosphorylation.
Collapse
Affiliation(s)
- Diogo R. Poroca
- Department of Physiology & BiophysicsDalhousie UniversityHalifaxNSCanada
| | - Noha Amer
- Department of Physiology & BiophysicsDalhousie UniversityHalifaxNSCanada
| | - Audrey Li
- Department of Physiology & BiophysicsDalhousie UniversityHalifaxNSCanada
| | | | - Valerie M. Chappe
- Department of Physiology & BiophysicsDalhousie UniversityHalifaxNSCanada
| |
Collapse
|