1
|
Schmidt SK, Fischer S, El Ahmad Z, Schmid R, Metzger E, Schüle R, Hellerbrand C, Arkudas A, Kengelbach-Weigand A, Kappelmann-Fenzl M, Bosserhoff AK. Modeling a mesenchymal cell state by bioprinting for the molecular analysis of dormancy in melanoma. Mater Today Bio 2025; 32:101674. [PMID: 40206148 PMCID: PMC11979991 DOI: 10.1016/j.mtbio.2025.101674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 04/11/2025] Open
Abstract
Malignant melanoma is a highly aggressive tumor originating from the pigment producing cells, the melanocytes. It accounts for the majority of skin cancer related deaths worldwide. This is often due to the development of therapy resistance or tumor dormancy, eventually resulting in tumor relapse by yet undefined mechanisms. Tumor dormancy is thought to be mediated by the cellular microenvironment and models taking this factor into account are urgently needed. We 3D bioprinted melanoma cells in the hydrogels Cellink Bioink (CIB) or Matrigel (MG), each as a substitute of the extracellular matrix, and, thereby, induced a quiescent or a proliferative phenotype of the melanoma cell lines, respectively. RNA-Seq with subsequent comprehensive bioinformatical and molecular analyses assigned CIB-cultured cells to a predominantly mesenchymal and Matrigel-cultured cells to a more mitotic phenotype, emphasizing the CIB model as a suitable platform for the investigation of dormancy under consideration of the microenvironment. Melanoma cells in CIB 3D culture reflect a quiescent and migratory active cell state e.g. by revealing significant downregulation of genes associated with replication and cell cycle progression in this setting. Using this model system, we identified the mechanosensory gene FHL2 as one early sensor of changes in the ECM and suggest a FHL2-p21/AP-1 axis contributing to the dormant phenotype of melanoma cells in CIB.
Collapse
Affiliation(s)
- Sonja K. Schmidt
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
| | - Stefan Fischer
- Faculty of Computer Science, Deggendorf Institute of Technology, Dieter-Görlitz-Platz 1, 94469, Deggendorf, Germany
| | - Zubeir El Ahmad
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
- Faculty of Computer Science, Deggendorf Institute of Technology, Dieter-Görlitz-Platz 1, 94469, Deggendorf, Germany
| | - Rafael Schmid
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Eric Metzger
- Klinik für Urologie und Zentrale Klinische Forschung, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-University Freiburg, 79106, Freiburg, Germany
| | - Roland Schüle
- Klinik für Urologie und Zentrale Klinische Forschung, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-University Freiburg, 79106, Freiburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
- CCC Erlangen-EMN: Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), 91054, Erlangen, Germany
- BZKF: Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany
| | - Andreas Arkudas
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Melanie Kappelmann-Fenzl
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
- Faculty of Computer Science, Deggendorf Institute of Technology, Dieter-Görlitz-Platz 1, 94469, Deggendorf, Germany
| | - Anja K. Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Fahrstraße 17, 91054, Erlangen, Germany
- CCC Erlangen-EMN: Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054, Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), 91054, Erlangen, Germany
- BZKF: Bavarian Cancer Research Center (BZKF), 91054, Erlangen, Germany
| |
Collapse
|
2
|
Man KF, Darweesh O, Hong J, Thompson A, O'Connor C, Bonaldo C, Melkonyan MN, Sun M, Patel R, Ellisen LW, Robinson T, Song D, Koh SB. CREB1-BCL2 drives mitochondrial resilience in RAS GAP-dependent breast cancer chemoresistance. Oncogene 2025; 44:1093-1105. [PMID: 39890967 PMCID: PMC11996675 DOI: 10.1038/s41388-025-03284-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogenous breast cancer subtype. RASAL2 is a RAS GTPase-activating protein (GAP) that has been associated with platinum resistance in TNBC, but the underlying mechanism is unknown. Here, we show that RASAL2 is enriched following neoadjuvant chemotherapy in TNBC patients. This enrichment is specific to the tumour compartment compared to adjacent normal tissues, suggesting that RASAL2 upregulation is tumour-selective. Analyses based on 2D/3D cultures and patient-derived xenograft models reveal that RASAL2 confers cross-resistance to common DNA-damaging chemotherapies other than platinum. Mechanistically, we found that apoptotic signalling is significantly downregulated upon RASAL2 expression. This feature is characterised by substantial alterations in the expression of anti-versus pro-apoptotic factors, pointing to heterogeneous mechanisms. In particular, RASAL2 upregulates BCL2 via activation of the oncogenic transcription co-factor YAP. CREB1, a YAP-interacting protein, was identified as the common transcription factor that binds to the promoter regions of RASAL2 and BCL2, driving their collective expression. A subset of RASAL2 colocalises with BCL2 subcellularly. Both proteins decorate mitochondria, where the high levels of mitochondrial RASAL2-induced BCL2 expression render the organelles refractory to apoptosis. Accordingly, mitochondrial outer membrane permeabilisation assay using live mitochondria from RASAL2-high/chemoresistant tumour cells demonstrated attenuated release of death signal, cytochrome c, when exposed to pro-apoptotic factors BAX and tBID. Similarly, these cells were more resilient towards chemotherapy-induced mitochondrial depolarisation. Together, this work reveals a previously undocumented molecular link between RAS GAP and apoptosis regulation, providing a new mechanistic framework for targeting a subset of chemorefractory tumours.
Collapse
Affiliation(s)
- Ki-Fong Man
- University of Bristol, University Walk, Bristol, UK
| | - Omeed Darweesh
- University of Bristol, University Walk, Bristol, UK
- College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
| | - Jinghui Hong
- University of Bristol, University Walk, Bristol, UK
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | - Mo Sun
- Department of Pathology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Rajnikant Patel
- University of Leicester, Henry Wellcome Building, Lancaster, UK
| | - Leif W Ellisen
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tim Robinson
- University of Bristol, University Walk, Bristol, UK
- University Hospitals Bristol and Weston, NHS Foundation Trust, Bristol, UK
| | - Dong Song
- Department of Breast Surgery, General Surgery Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Siang-Boon Koh
- University of Bristol, University Walk, Bristol, UK.
- University Hospitals Bristol and Weston, NHS Foundation Trust, Bristol, UK.
| |
Collapse
|
3
|
Kalyoncu M, Demirci D, Eris S, Dayanc B, Cakiroglu E, Basol M, Uysal M, Cakan-Akdogan G, Liu F, Ozturk M, Karakülah G, Senturk S. Escape from TGF-β-induced senescence promotes aggressive hallmarks in epithelial hepatocellular carcinoma cells. Mol Oncol 2025. [PMID: 40083231 DOI: 10.1002/1878-0261.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 01/16/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Transforming growth factor-β (TGF-β) signaling and cellular senescence are key hallmarks of hepatocellular carcinoma (HCC) pathogenesis. Despite provoking senescence-associated growth arrest in epithelial HCC cells, elevated TGF-β activity paradoxically correlates with increased aggressiveness and poor prognosis in advanced tumors. Whether the transition between these dichotomous functions involves modulation of the senescence phenotype during disease progression remains elusive. Exploiting the epithelial HCC cell line Huh7 as a robust model, we demonstrate that chronic exposure to TGF-β prompts escape from Smad3-mediated senescence, leading to the development of TGF-β resistance. This altered state is characterized by an optimal proliferation rate and the acquisition of molecular and functional traits of less-differentiated mesenchymal cells, coinciding with differential growth capacity in 2D and 3D culture conditions, epithelial-to-mesenchymal transition (EMT), and increased invasiveness in vitro, and metastasis in vivo. Mechanistically, resistant cells exhibit defective activation and nuclear trafficking of Smad molecules, particularly Smad3, as ectopic activation of the TGF-β/Smad3 axis is able to reinstate TGF-β sensitivity. An integrated transcriptomic landscape reveals both shared and distinct gene signatures associated with senescent and TGF-β resistant states. Importantly, genetic ablation and molecular studies identify microtubule affinity regulating kinase 1 (MARK1) and glutamate metabotropic receptor 8 (GRM8) as critical modulators of the resistance phenomenon, potentially by impairing spatiotemporal signaling dynamics of Smad activity. Our findings unveil a novel phenomenon wherein epithelial HCC cells may exploit senescence plasticity as a mechanism to oppose TGF-β anti-tumor responses and progress towards more aggressive HCC phenotypes.
Collapse
Affiliation(s)
| | | | - Sude Eris
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Bengisu Dayanc
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ece Cakiroglu
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Merve Basol
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Merve Uysal
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Gulcin Cakan-Akdogan
- Izmir Biomedicine and Genome Center, Turkey
- Department of Biomedicine and Health Technologies, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Susan Lehman Cullman Laboratory for Cancer Research, Ernest Mario School of Pharmacy, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Mehmet Ozturk
- Department of Medical Biology, Izmir Tinaztepe University School of Medicine, Turkey
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Gökhan Karakülah
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Turkey
- Department of Genomics and Molecular Biotechnology, Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| |
Collapse
|
4
|
Yang S, Xue B, Zhang Y, Wu H, Yu B, Li S, Ma T, Gao X, Hao Y, Guo L, Liu Q, Gao X, Yang Y, Wang Z, Qin M, Tian Y, Fu L, Zhou B, Li L, Li J, Gong S, Xia B, Huang J. Engineered Extracellular Vesicles from Antler Blastema Progenitor Cells: A Therapeutic Choice for Spinal Cord Injury. ACS NANO 2025; 19:5995-6013. [PMID: 39841785 PMCID: PMC11841045 DOI: 10.1021/acsnano.4c10298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/24/2025]
Abstract
Deer antler blastema progenitor cells (ABPCs) are promising for regenerative medicine due to their role in annual antler regeneration, the only case of complete organ regeneration in mammals. ABPC-derived signals show great potential for promoting regeneration in tissues with limited natural regenerative ability. Our findings demonstrate the capability of extracellular vesicles from ABPCs (EVsABPC) to repair spinal cord injury (SCI), a condition with low regenerative capacity. EVsABPC significantly enhanced the proliferation of neural stem cells (NSCs) and activated neuronal regenerative potential, resulting in a 5.2-fold increase in axonal length. Additionally, EVsABPC exhibited immunomodulatory effects, shifting macrophages from M1 to M2. Engineered with activated cell-penetrating peptides (ACPPs), EVsABPC significantly outperformed EVs from rat bone marrow stem cells (EVsBMSC) and neural stem cells (EVsNSC), promoting a 1.3-fold increase in axonal growth, a 30.6% reduction in neuronal apoptosis, and a 2.6-fold improvement in motor function recovery. These findings support ABPC-derived EVs as a promising therapeutic candidate for SCI repair.
Collapse
Affiliation(s)
- Shijie Yang
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Borui Xue
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
- Air
Force 986(th) Hospital, The Fourth Military
Medical University, Xi’an 710001, P.R. China
| | - Yongfeng Zhang
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Haining Wu
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Beibei Yu
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Shengyou Li
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Teng Ma
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Xue Gao
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Yiming Hao
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Lingli Guo
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Qi Liu
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Xueli Gao
- School
of
Ecology and Environment, Northwestern Polytechnical
University, Xi’an 710072, P.R. China
| | - Yujie Yang
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Zhenguo Wang
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Mingze Qin
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Yunze Tian
- Department
of Thoracic Surgery, Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Longhui Fu
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Bisheng Zhou
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Luyao Li
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Jianzhong Li
- Department
of Thoracic Surgery, Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
| | - Shouping Gong
- Department
of Neurosurgery, The Second Affiliated Hospital
of Xi’an Jiao Tong University, Xi’an 710004, P.R. China
- Xi’an
Medical University, Xi’an 710021, P.R. China
| | - Bing Xia
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| | - Jinghui Huang
- Department
of Orthopaedics, Xijing Hospital, The Fourth
Military Medical University, Xi’an 710032, P.R. China
| |
Collapse
|
5
|
Chen H, Gou X, Mao Y, O'Leary PC, Diolaiti ME, Ashworth A. PARP7 Inhibitors and AHR Agonists Act Synergistically across a Wide Range of Cancer Models. Mol Cancer Ther 2025; 24:56-68. [PMID: 39313957 DOI: 10.1158/1535-7163.mct-24-0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/13/2024] [Accepted: 09/11/2024] [Indexed: 09/25/2024]
Abstract
Small-molecule inhibitors of the mono (ADP) ribosyl transferase PARP7 are being evaluated asmonotherapy for tumors overexpressing PARP7 and in combination with immune checkpoint blockade. We previously showed that sensitivity to the PARP7 inhibitor (PARP7i) RBN-2397 could be enhanced by cotreatment with agonists of the aryl hydrocarbon receptor (AHRa) in cell lines that show strong intrinsic sensitivity to RBN-2397. In this study, we demonstrated that a range of tumor cell lines that are relatively insensitive to PARP7i or AHRa as individual agents are unexpectedly profoundly sensitive to their combination. Our data show that this synergistic response is dependent on the AHR/AHR nuclear translocator and is associated with increased levels of nuclear AHR and increased transcription of AHR target genes. In some hormone receptor-positive cell lines, we find that combination treatment is associated with proteasomal turnover of the steroid hormone receptors, androgen receptor and estrogen receptor. Both wild-type and hormone-resistant mutant forms of these receptors are degraded upon treatment with AHRa and PARP7i in breast and prostate cancer models. These results suggest that combining PARP7i with AHRa may extend the utility of these drugs to a wider range of tumors, including those that are refractory to hormone therapy.
Collapse
Affiliation(s)
- Huadong Chen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Xuxu Gou
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Ying Mao
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Patrick C O'Leary
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Morgan E Diolaiti
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California
| |
Collapse
|
6
|
Needham JM, Perritt SE, Thompson SR. Single-cell analysis reveals host S phase drives large T antigen expression during BK polyomavirus infection. PLoS Pathog 2024; 20:e1012663. [PMID: 39636788 PMCID: PMC11620372 DOI: 10.1371/journal.ppat.1012663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
BK polyomavirus (BKPyV) is a major cause of kidney transplant failure, for which there are no antivirals. The current model is that BKPyV expresses TAg (large T antigen) early during infection, promoting cells to enter S phase where the viral DNA can access the host replication machinery. Here, we performed a single-cell analysis of viral TAg expression throughout the cell cycle to reveal that robust TAg expression required replication of the host DNA first. By using inhibitors that only affect host and not viral replication, we show that both TAg expression and viral production rely on an initial S phase. BKPyV is known to promote cellular re-replication, where the cell re-enters S phase from G2 phase (without passing through mitosis or G1 phase) to prolong S phase for viral replication. Thus, BKPyV infection results in cells with greater than 4N DNA content. We found that these subsequent rounds of replication of the host DNA relied on canonical host cell cycle machinery and regulators despite BKPyV infection. Together, these findings suggest a model for polyomavirus replication, where robust viral TAg expression depends on an initial host S phase and that BKPyV primarily replicates during host re-replication. Having a better understanding of the molecular events that are required for BKPyV production will help identify effective therapeutic targets against BKPyV.
Collapse
Affiliation(s)
- Jason M. Needham
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama United States of America
| | - Sarah E. Perritt
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama United States of America
| | - Sunnie R. Thompson
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama United States of America
| |
Collapse
|
7
|
Ay M, Veleva D, Chowdhury M, Drljaca S, Sobanski T, Prowse ABJ, Ovchinnikov DA. An inducible iFUCCI reporter cell line for tracking cell cycle dynamics in human pluripotent stem cells and their differentiated progeny. Stem Cell Res 2024; 81:103614. [PMID: 39571285 DOI: 10.1016/j.scr.2024.103614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/15/2024] Open
Abstract
An ability to monitor cell cycle progression in real time has numerous applications in pluripotent stem cell-based models and therapeutics development. Fluorescence Ubiquitination-based Cell Cycle Indicator (FUCCI) systems enable live monitoring of the cell cycle in stem cells and their differentiated progenies in a non-invasive manner. We describe the generation and characterisation of a cell line with a doxycycline-inducible reporter system, iFUCCI, in the human embryonic stem cell (hESC) line MEL-1. MEL-1 iFUCCI hESCs exhibited a normal karyotype, expressed markers of the undifferentiated state, and demonstrated expected differentiation potential by giving rise to cell populations from all three germ layers.
Collapse
Affiliation(s)
- Merve Ay
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Desi Veleva
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia.
| | - Mohammad Chowdhury
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Sonja Drljaca
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Thais Sobanski
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Andrew B J Prowse
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia
| | - Dmitry A Ovchinnikov
- StemCore, The Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia 4072, QLD, Australia; The Florey, The University of Melbourne, 30 Royal Parade, Melbourne, VIC 3010, Australia
| |
Collapse
|
8
|
Hayatigolkhatmi K, Soriani C, Soda E, Ceccacci E, El Menna O, Peri S, Negrelli I, Bertolini G, Franchi GM, Carbone R, Minucci S, Rodighiero S. Automated workflow for the cell cycle analysis of (non-)adherent cells using a machine learning approach. eLife 2024; 13:RP94689. [PMID: 39576677 PMCID: PMC11584176 DOI: 10.7554/elife.94689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024] Open
Abstract
Understanding the cell cycle at the single-cell level is crucial for cellular biology and cancer research. While current methods using fluorescent markers have improved the study of adherent cells, non-adherent cells remain challenging. In this study, we addressed this gap by combining a specialized surface to enhance cell attachment, the FUCCI(CA)2 sensor, an automated image analysis pipeline, and a custom machine learning algorithm. This approach enabled precise measurement of cell cycle phase durations in non-adherent cells. This method was validated in acute myeloid leukemia cell lines NB4 and Kasumi-1, which have unique cell cycle characteristics, and we tested the impact of cell cycle-modulating drugs on NB4 cells. Our cell cycle analysis system, which is also compatible with adherent cells, is fully automated and freely available, providing detailed insights from hundreds of cells under various conditions. This report presents a valuable tool for advancing cancer research and drug development by enabling comprehensive, automated cell cycle analysis in both adherent and non-adherent cells.
Collapse
Affiliation(s)
| | - Chiara Soriani
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| | - Emanuel Soda
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| | - Elena Ceccacci
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| | - Oualid El Menna
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| | - Sebastiano Peri
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| | | | | | | | | | - Saverio Minucci
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
- Department of Oncology and Hemato-Oncology, University of MilanMilanItaly
| | - Simona Rodighiero
- Department of Experimental Oncology, European Institute of Oncology-IRCCSMilanItaly
| |
Collapse
|
9
|
Nerger BA, Kashyap K, Deveney BT, Lou J, Hanan BF, Liu Q, Khalil A, Lungjangwa T, Cheriyan M, Gupta A, Jaenisch R, Weitz DA, Mahadevan L, Mooney DJ. Tuning porosity of macroporous hydrogels enables rapid rates of stress relaxation and promotes cell expansion and migration. Proc Natl Acad Sci U S A 2024; 121:e2410806121. [PMID: 39467139 PMCID: PMC11551365 DOI: 10.1073/pnas.2410806121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Extracellular matrix (ECM) viscoelasticity broadly regulates cell behavior. While hydrogels can approximate the viscoelasticity of native ECM, it remains challenging to recapitulate the rapid stress relaxation observed in many tissues without limiting the mechanical stability of the hydrogel. Here, we develop macroporous alginate hydrogels that have an order of magnitude increase in the rate of stress relaxation as compared to bulk hydrogels. The increased rate of stress relaxation occurs across a wide range of polymer molecular weights (MWs), which enables the use of high MW polymer for improved mechanical stability of the hydrogel. The rate of stress relaxation in macroporous hydrogels depends on the volume fraction of pores and the concentration of bovine serum albumin, which is added to the hydrogels to stabilize the macroporous structure during gelation. Relative to cell spheroids encapsulated in bulk hydrogels, spheroids in macroporous hydrogels have a significantly larger area and smaller circularity because of increased cell migration. A computational model provides a framework for the relationship between the macroporous architecture and morphogenesis of encapsulated spheroids that is consistent with experimental observations. Taken together, these findings elucidate the relationship between macroporous hydrogel architecture and stress relaxation and help to inform the design of macroporous hydrogels for materials-based cell therapies.
Collapse
Affiliation(s)
- Bryan A. Nerger
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Kirti Kashyap
- Department of Physics, Indian Institute of Technology Hyderabad, Sangareddy, Telangana502285, India
| | - Brendan T. Deveney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Junzhe Lou
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Blake F. Hanan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| | - Qi Liu
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | - Andrew Khalil
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
| | | | | | - Anupam Gupta
- Department of Physics, Indian Institute of Technology Hyderabad, Sangareddy, Telangana502285, India
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02142
| | - David A. Weitz
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Physics, Harvard University, Cambridge, MA02138
| | - L. Mahadevan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Department of Physics, Harvard University, Cambridge, MA02138
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA02138
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
| |
Collapse
|
10
|
Xue B, Peng Y, Zhang Y, Yang S, Zheng Y, Hu H, Gao X, Yu B, Gao X, Li S, Wu H, Ma T, Hao Y, Wei Y, Guo L, Yang Y, Wang Z, Xue T, Zhang J, Luo B, Xia B, Huang J. A Novel Superparamagnetic-Responsive Hydrogel Facilitates Disc Regeneration by Orchestrating Cell Recruitment, Proliferation, and Differentiation within Hostile Inflammatory Niche. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408093. [PMID: 39373392 PMCID: PMC11600201 DOI: 10.1002/advs.202408093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/21/2024] [Indexed: 10/08/2024]
Abstract
In situ disc regeneration is a meticulously orchestrated process, which involves cell recruitment, proliferation and differentiation within a local inflammatory niche. Thus far, it remains a challenge to establish a multi-staged regulatory framework for coordinating these cellular events, therefore leading to unsatisfactory outcome. This study constructs a super paramagnetically-responsive cellular gel, incorporating superparamagnetic iron oxide nanoparticles (SPIONs) and aptamer-modified palladium-hydrogen nanozymes (PdH-Apt) into a double-network polyacrylamide/hyaluronic acid (PAAm/HA) hydrogel. The Aptamer DB67 within magnetic hydrogel (Mag-gel) showed a high affinity for disialoganglioside (GD2), a specific membrane ligand of nucleus pulposus stem cells (NPSCs), to precisely recruit them to the injury site. The Mag-gel exhibits remarkable sensitivity to a magnetic field (MF), which exerts tunable micro/nano-scale forces on recruited NPSCs and triggers cytoskeletal remodeling, consequently boosting cell expansion in the early stage. By altering the parameters of MF, the mechanical cues within the hydrogel facilitates differentiation of NPSCs into nucleus pulposus cells to restore disc structure in the later stage. Furthermore, the PdH nanozymes within the Mag-gel mitigate the harsh inflammatory microenvironment, favoring cell survival and disc regeneration. This study presents a remote and multi-staged strategy for chronologically regulating endogenous stem cell fate, supporting disc regeneration without invasive procedures.
Collapse
Affiliation(s)
- Borui Xue
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
- Air Force 986(th) HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Yan Peng
- College of Advanced ManufacturingFuzhou UniversityJinjiang362200P. R. China
| | - Yongfeng Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Shijie Yang
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Yi Zheng
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Huiling Hu
- Air Force 986(th) HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Xueli Gao
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Beibei Yu
- Department of NeurosurgeryThe Second Affiliated Hospital of Xi'an Jiao Tong UniversityXi'an710032P. R. China
| | - Xue Gao
- School of Ecology and EnvironmentNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Shengyou Li
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Haining Wu
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Teng Ma
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Yiming Hao
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Yitao Wei
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Lingli Guo
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Yujie Yang
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Zhenguo Wang
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Tingfeng Xue
- School of Ecology and EnvironmentNorthwestern Polytechnical UniversityXi'an710072P. R. China
| | - Jin Zhang
- College of Chemical EngineeringFuzhou UniversityXueyuan RoadFuzhou350108P. R. China
| | - Beier Luo
- Department of Spinal SurgeryShanghai Changhai HospitalAffiliated to Naval Medical UniversityShanghai200433P. R. China
| | - Bing Xia
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Jinghui Huang
- Department of OrthopaedicsXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| |
Collapse
|
11
|
Ecker A, Lázár B, Tóth RI, Urbán M, Hoffmann OI, Fekete Z, Barta E, Uher F, Matula Z, Várkonyi E, Gócza E. Creating a novel method for chicken primordial germ cell health monitoring using the fluorescent ubiquitination-based cell cycle indicator reporter system. Poult Sci 2024; 103:104144. [PMID: 39173570 PMCID: PMC11382113 DOI: 10.1016/j.psj.2024.104144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
The most current in vitro genetic methods, including gene preservation, gene editing and developmental modelling, require a significant number of healthy cells. In poultry species, primordial germ cells (PGCs) are great candidates for all the above-mentioned purposes, given their easy culturing and well-established freezing method for chicken. However, the constant monitoring of cultures can be financially challenging and consumes large amounts of solutions and accessories. This study aimed to introduce the Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI) complex into the chicken PGCs. FUCCI is a powerful transgenic tool based on the periodic protein expression changes during the cell cycle. It includes chromatin licensing and DNA replication factor 1 attached monomeric Kusabira-Orange and Geminin-attached monomeric Azami-Green fluorescent proteins, that cause the cells to express a red signal in the G1 phase and a green signal in S and G2 phases. Modification of the chicken PGCs was done via electroporation and deemed to be successful according to confocal microscopy, DNA sequencing and timelapse video analysis. Stable clone cell lines were established, cryopreserved, and injected into recipient embryos to prove the integrational competency. The cell health monitoring was tested with medium change experiments, that proved the intended reactions of the FUCCI transgene. These results established the future for FUCCI experiments in chicken, including heat treatment and toxin treatment.
Collapse
Affiliation(s)
- András Ecker
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Bence Lázár
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary; National Centre for Biodiversity and Gene Conservation, Gödöllő, 2100 Hungary
| | - Roland I Tóth
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Martin Urbán
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Orsolya I Hoffmann
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Zsófia Fekete
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, 80101 Finland
| | - Endre Barta
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, 4032 Hungary
| | - Ferenc Uher
- National Institute of Hematology and Infectology, Budapest, 1097 Hungary
| | - Zsolt Matula
- National Institute of Hematology and Infectology, Budapest, 1097 Hungary
| | - Eszter Várkonyi
- National Centre for Biodiversity and Gene Conservation, Gödöllő, 2100 Hungary
| | - Elen Gócza
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary.
| |
Collapse
|
12
|
Mehta S, Buyanbat A, Kai Y, Orkin SH. Mechanistic and kinetic insights into transcription factor biology via acute protein depletion. Exp Hematol 2024; 137:104250. [PMID: 38862078 DOI: 10.1016/j.exphem.2024.104250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/13/2024]
Abstract
Genetic downregulation of the BCL11A transcription factor (TF) reverses the switch from fetal to adult hemoglobin and is effective in treating β-hemoglobinopathies. Genetic ablation results in a gradual reduction in protein abundance and does not lend itself to the analysis of the immediate consequences of protein loss or the determination of the direct interactors/targets of the protein of interest. We achieved acute degradation of the largely disordered and 'undruggable' BCL11A protein by fusing it with a conditional degradation (degron) tag, FKBP12F36V, called degradable tags (dTAG). Small molecules then depleted the BCL11A-dTAG through endogenous proteolytic pathways. By integrating acute depletion with nascent transcriptomics and cell cycle separation techniques, we demonstrate the necessity of BCL11A occupancy at the target chromatin for sustained transcriptional repression in erythroid cells. We advocate for expanding the exploration of TF function to include acute depletion, which holds the potential to unveil unprecedented kinetic insights into TF mechanisms of action.
Collapse
Affiliation(s)
- Stuti Mehta
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Altantsetseg Buyanbat
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Yan Kai
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Stuart H Orkin
- Dana Farber/Boston Children's Hospital Cancer and Blood Disorder Center, Department of Pediatrics, Harvard Medical School, Boston, MA; Boston Children's Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA.
| |
Collapse
|
13
|
Niu Y, Luo J, Zong C. Single-cell total-RNA profiling unveils regulatory hubs of transcription factors. Nat Commun 2024; 15:5941. [PMID: 39009595 PMCID: PMC11251146 DOI: 10.1038/s41467-024-50291-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Recent development of RNA velocity uses master equations to establish the kinetics of the life cycle of RNAs from unspliced RNA to spliced RNA (i.e., mature RNA) to degradation. To feed this kinetic analysis, simultaneous measurement of unspliced RNA and spliced RNA in single cells is greatly desired. However, the majority of single-cell RNA-seq chemistry primarily captures mature RNA species to measure gene expressions. Here, we develop a one-step total-RNA chemistry-based single-cell RNA-seq method: snapTotal-seq. We benchmark this method with multiple single-cell RNA-seq assays in their performance in kinetic analysis of cell cycle by RNA velocity. Next, with LASSO regression between transcription factors, we identify the critical regulatory hubs mediating the cell cycle dynamics. We also apply snapTotal-seq to profile the oncogene-induced senescence and identify the key regulatory hubs governing the entry of senescence. Furthermore, from the comparative analysis of unspliced RNA and spliced RNA, we identify a significant portion of genes whose expression changes occur in spliced RNA but not to the same degree in unspliced RNA, indicating these gene expression changes are mainly controlled by post-transcriptional regulation. Overall, we demonstrate that snapTotal-seq can provide enriched information about gene regulation, especially during the transition between cell states.
Collapse
Affiliation(s)
- Yichi Niu
- Department of Molecular and Human Genetics, Houston, TX, USA
- Genetics & Genomics Program, Houston, TX, USA
| | - Jiayi Luo
- Department of Molecular and Human Genetics, Houston, TX, USA
- Cancer and Cell Biology Program, Houston, TX, USA
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Houston, TX, USA.
- Genetics & Genomics Program, Houston, TX, USA.
- Cancer and Cell Biology Program, Houston, TX, USA.
- Integrative Molecular and Biomedical Sciences Program, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Gillespie W, Zhang Y, Ruiz OE, Cerda J, Ortiz-Guzman J, Turner WD, Largoza G, Sherman M, Mosser LE, Fujimoto E, Chien CB, Kwan KM, Arenkiel BR, Devine WP, Wythe JD. Multisite Assembly of Gateway Induced Clones (MAGIC): a flexible cloning toolbox with diverse applications in vertebrate model systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603267. [PMID: 39026881 PMCID: PMC11257631 DOI: 10.1101/2024.07.13.603267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Here we present the Multisite Assembly of Gateway Induced Clones (MAGIC) system, which harnesses site-specific recombination-based cloning via Gateway technology for rapid, modular assembly of between 1 and 3 "Entry" vector components, all into a fourth, standard high copy "Destination" plasmid backbone. The MAGIC toolkit spans a range of in vitro and in vivo uses, from directing tunable gene expression, to driving simultaneous expression of microRNAs and fluorescent reporters, to enabling site-specific recombinase-dependent gene expression. All MAGIC system components are directly compatible with existing multisite gateway Tol2 systems currently used in zebrafish, as well as existing eukaryotic cell culture expression Destination plasmids, and available mammalian lentiviral and adenoviral Destination vectors, allowing rapid cross-species experimentation. Moreover, herein we describe novel vectors with flanking piggyBac transposon elements for stable genomic integration in vitro or in vivo when used with piggyBac transposase. Collectively, the MAGIC system facilitates transgenesis in cultured mammalian cells, electroporated mouse and chick embryos, as well as in injected zebrafish embryos, enabling the rapid generation of innovative DNA constructs for biological research due to a shared, common plasmid platform.
Collapse
|
15
|
Schipka R, Heltmann-Meyer S, Schneidereit D, Friedrich O, Röder J, Boccaccini AR, Schrüfer S, Schubert DW, Horch RE, Bosserhoff AK, Arkudas A, Kengelbach-Weigand A, Schmid R. Characterization of two different alginate-based bioinks and the influence of melanoma growth within. Sci Rep 2024; 14:12945. [PMID: 38839791 PMCID: PMC11153560 DOI: 10.1038/s41598-024-63642-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024] Open
Abstract
Extrusion-based bioprinting is an established method in biofabrication. Suitable bioinks have fundamentally different compositions and characteristics, which should be examined, in order to find a perfect model system. Here, we investigate the effect of two alginate-based, yet unalike 3D-printed bioinks, pre-crosslinked alginate-dialdehyde gelatin (ADA-GEL) and a mixture of alginate, hyaluronic acid, and gelatin (Alg/HA/Gel), on the melanoma cell line Mel Im and vice versa in terms of stiffness, shrinkage, cellular behavior and colony formation over 15 days. Rheological stiffness measurements revealed two soft gels with similar storage moduli. The cells did not have a significant impact on the overall stiffness, whereas ADA-GEL (2.5/2.5%) was significantly stiffer than Alg/HA/Gel (0.5/0.1/3%). Regarding the shrinkage of printed constructs, cells had a significant influence, especially in ADA-GEL, which has covalent bonds between the oxidized alginate and gelatin. Multi-photon microscopy exhibited proliferation, cell spreading and migration in ADA-GEL with cell-cell and cell-matrix interaction, dissimilarly to Alg/HA/Gel, in which cells formed spherical, encapsulated colonies. Scanning electron microscopy and histology showed degradation and multi-layered growth on ADA-GEL and fewer examples of escaped cells on Alg/HA/Gel. Both gels serve as proliferation bioink for melanoma with more necrosis in deeper Alg/HA/Gel colonies and differences in spreading and matrix interaction. These findings show the importance of proper characterization of the bioinks for different applications.
Collapse
Affiliation(s)
- Raphael Schipka
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Stefanie Heltmann-Meyer
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Jonas Röder
- Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
- RevoBITs, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Rafael Schmid
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany.
| |
Collapse
|
16
|
Chowdhury SP, Solley SC, Polishchuk E, Bacal J, Conrad JE, Gardner BM, Acosta-Alvear D, Zappa F. Baseline unfolded protein response signaling adjusts the timing of the mammalian cell cycle. Mol Biol Cell 2024; 35:br12. [PMID: 38656789 PMCID: PMC11238080 DOI: 10.1091/mbc.e23-11-0419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024] Open
Abstract
The endoplasmic reticulum (ER) is a single-copy organelle that cannot be generated de novo, suggesting coordination between the mechanisms overseeing ER integrity and those controlling the cell cycle to maintain organelle inheritance. The Unfolded Protein Response (UPR) is a conserved signaling network that regulates ER homeostasis. Here, we show that pharmacological and genetic inhibition of the UPR sensors IRE1, ATF6, and PERK in unstressed cells delays the cell cycle, with PERK inhibition showing the most penetrant effect, which was associated with a slowdown of the G1-to-S/G2 transition. Treatment with the small molecule ISRIB to bypass the effects of PERK-dependent phosphorylation of the translation initiation factor eIF2α had no such effect, suggesting that cell cycle timing depends on PERK's kinase activity but is independent of eIF2α phosphorylation. Using complementary light and electron microscopy and flow cytometry-based analyses, we also demonstrate that the ER enlarges before mitosis. Together, our results suggest coordination between UPR signaling and the cell cycle to maintain ER physiology during cell division.
Collapse
Affiliation(s)
- Soham P. Chowdhury
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Sabrina C. Solley
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Elena Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
| | - Julien Bacal
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Julia E. Conrad
- Altos Labs Bay Area Institute of Science, Altos Labs, Redwood City, CA 94065
| | - Brooke M. Gardner
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Diego Acosta-Alvear
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| | - Francesca Zappa
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106
| |
Collapse
|
17
|
Schneider MP, Cullen AE, Pangonyte J, Skelton J, Major H, Van Oudenhove E, Garcia MJ, Chaves Urbano B, Piskorz AM, Brenton JD, Macintyre G, Markowetz F. scAbsolute: measuring single-cell ploidy and replication status. Genome Biol 2024; 25:62. [PMID: 38438920 PMCID: PMC10910719 DOI: 10.1186/s13059-024-03204-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
Cancer cells often exhibit DNA copy number aberrations and can vary widely in their ploidy. Correct estimation of the ploidy of single-cell genomes is paramount for downstream analysis. Based only on single-cell DNA sequencing information, scAbsolute achieves accurate and unbiased measurement of single-cell ploidy and replication status, including whole-genome duplications. We demonstrate scAbsolute's capabilities using experimental cell multiplets, a FUCCI cell cycle expression system, and a benchmark against state-of-the-art methods. scAbsolute provides a robust foundation for single-cell DNA sequencing analysis across different technologies and has the potential to enable improvements in a number of downstream analyses.
Collapse
Affiliation(s)
- Michael P Schneider
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Amy E Cullen
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Justina Pangonyte
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Jason Skelton
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Harvey Major
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Elke Van Oudenhove
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Maria J Garcia
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Anna M Piskorz
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - James D Brenton
- University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK
| | - Geoff Macintyre
- Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Florian Markowetz
- University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, Robinson Way, Cambridge, UK.
| |
Collapse
|
18
|
Korsnes MS, Korsnes R. Initial refinement of data from video-based single-cell tracking. CANCER INNOVATION 2023; 2:416-432. [PMID: 38090384 PMCID: PMC10686135 DOI: 10.1002/cai2.88] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/13/2023] [Accepted: 06/01/2023] [Indexed: 10/15/2024]
Abstract
Background Video recording of cells offers a straightforward way to gain valuable information from their response to treatments. An indispensable step in obtaining such information involves tracking individual cells from the recorded data. A subsequent step is reducing such data to represent essential biological information. This can help to compare various single-cell tracking data yielding a novel source of information. The vast array of potential data sources highlights the significance of methodologies prioritizing simplicity, robustness, transparency, affordability, sensor independence, and freedom from reliance on specific software or online services. Methods The provided data presents single-cell tracking of clonal (A549) cells as they grow in two-dimensional (2D) monolayers over 94 hours, spanning several cell cycles. The cells are exposed to three different concentrations of yessotoxin (YTX). The data treatments showcase the parametrization of population growth curves, as well as other statistical descriptions. These include the temporal development of cell speed in family trees with and without cell death, correlations between sister cells, single-cell average displacements, and the study of clustering tendencies. Results Various statistics obtained from single-cell tracking reveal patterns suitable for data compression and parametrization. These statistics encompass essential aspects such as cell division, movements, and mutual information between sister cells. Conclusion This work presents practical examples that highlight the abundant potential information within large sets of single-cell tracking data. Data reduction is crucial in the process of acquiring such information which can be relevant for phenotypic drug discovery and therapeutics, extending beyond standardized procedures. Conducting meaningful big data analysis typically necessitates a substantial amount of data, which can stem from standalone case studies as an initial foundation.
Collapse
Affiliation(s)
- Mónica Suárez Korsnes
- Department of Clinical and Molecular MedicineNorwegian University of Science and Technology (NTNU)TrondheimNorway
- Korsnes Biocomputing (KoBio)TrondheimNorway
| | | |
Collapse
|
19
|
Maeta K, Farea M, Nishio H, Matsuo M. A novel splice variant of the human MSTN gene encodes a myostatin-specific myostatin inhibitor. J Cachexia Sarcopenia Muscle 2023; 14:2289-2300. [PMID: 37582652 PMCID: PMC10570081 DOI: 10.1002/jcsm.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/02/2022] [Accepted: 07/11/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Myostatin, encoded by the MSTN gene comprising 3 exons, is a potent negative regulator of skeletal muscle growth. Although a variety of myostatin inhibitors have been invented for increasing muscle mass in muscle wasting diseases, no effective inhibitor is currently available for clinical use. Myostatin isoforms in several animals have been reported to inhibit myostatin, but an isoform has never been identified for the human MSTN gene, a conserved gene among animals. Here, a splice variant of the human MSTN gene was explored. METHODS Transcripts and proteins were analysed by reverse transcription-PCR amplification and western blotting, respectively. Proteins were expressed from expression plasmid. Myostatin signalling was assayed by the SMAD-responsive luciferase activity. Cell proliferation was assayed by the Cell Counting Kit-8 (CCK-8) assay and cell counting. Cell cycle was analysed by the FastFUCCI system. RESULTS Reverse transcription-PCR amplification of the full-length MSTN transcript in CRL-2061 rhabdomyosarcoma cells revealed two bands consisting of a thick expected-size product and a thin additional small-size product. Sequencing of the small-size product showed a 963-bp deletion in the 5' end of exon 3, creating exon 3s, which contained unusual splice acceptor TG dinucleotides. The novel variant was identified in other human cell lines, although it was not identified in skeletal muscle. The 251-amino acid isoform encoded by the novel variant (myostatin-b) was identified in CRL-2061 rhabdomyosarcoma cells. Transfection of a myostatin-b expression plasmid into CRL-2061 and myoblast cells inhibited endogenous myostatin signalling (44%, P < 0.001 and 63%, P < 0.001, respectively). Furthermore, myostatin-b inhibited myostatin signalling induced by recombinant myostatin (68.8%, P < 0.001). In remarkable contrast, myostatin-b did not inhibit the myostatin signalling induced by recombinant growth differentiation factor 11 (9.2%, P = 0.70), transforming growth factor β (+3.1%, P = 0.83) or activin A (+1.1%, P = 0.96). These results indicate the myostatin-specific inhibitory effect of myostatin-b. Notably, the expression of myostatin-b in myoblasts significantly enhanced cell proliferation higher than the mock-transfected cells by the CCK-8 and direct cell counting assays (60%, P < 0.05 and 39%, P < 0.05, respectively). Myostatin-b increased the percentage of S-phase cells significantly higher than that of the mock-transfected cells (53% vs. 80%, P < 0.05). CONCLUSIONS We cloned a novel human MSTN variant produced by unorthodox splicing. The variant encoded a novel myostatin isoform, myostatin-b, that inhibited myostatin signalling by myostatin-specific manner and enhanced myoblast proliferation by shifting cell cycle. Myostatin-b, which has myostatin-specific inhibitory activity, could be developed as a natural myostatin inhibitor.
Collapse
Affiliation(s)
- Kazuhiro Maeta
- KNC Department of Nucleic Acid Drug Discovery, Faculty of RehabilitationKobe Gakuin UniversityKobeJapan
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
| | - Manal Farea
- KNC Department of Nucleic Acid Drug Discovery, Faculty of RehabilitationKobe Gakuin UniversityKobeJapan
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
| | - Hisahide Nishio
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
- Department of Occupational Therapy, Faculty of RehabilitationKobe Gakuin UniversityKobeJapan
| | - Masafumi Matsuo
- KNC Department of Nucleic Acid Drug Discovery, Faculty of RehabilitationKobe Gakuin UniversityKobeJapan
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
| |
Collapse
|
20
|
Acharya BR, Fang JS, Jeffery ED, Chavkin NW, Genet G, Vasavada H, Nelson EA, Sheynkman GM, Humphries MJ, Hirschi KK. Connexin 37 sequestering of activated-ERK in the cytoplasm promotes p27-mediated endothelial cell cycle arrest. Life Sci Alliance 2023; 6:e202201685. [PMID: 37197981 PMCID: PMC10192821 DOI: 10.26508/lsa.202201685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023] Open
Abstract
Connexin37-mediated regulation of cell cycle modulators and, consequently, growth arrest lack mechanistic understanding. We previously showed that arterial shear stress up-regulates Cx37 in endothelial cells and activates a Notch/Cx37/p27 signaling axis to promote G1 cell cycle arrest, and this is required to enable arterial gene expression. However, how induced expression of a gap junction protein, Cx37, up-regulates cyclin-dependent kinase inhibitor p27 to enable endothelial growth suppression and arterial specification is unclear. Herein, we fill this knowledge gap by expressing wild-type and regulatory domain mutants of Cx37 in cultured endothelial cells expressing the Fucci cell cycle reporter. We determined that both the channel-forming and cytoplasmic tail domains of Cx37 are required for p27 up-regulation and late G1 arrest. Mechanistically, the cytoplasmic tail domain of Cx37 interacts with, and sequesters, activated ERK in the cytoplasm. This then stabilizes pERK nuclear target Foxo3a, which up-regulates p27 transcription. Consistent with previous studies, we found this Cx37/pERK/Foxo3a/p27 signaling axis functions downstream of arterial shear stress to promote endothelial late G1 state and enable up-regulation of arterial genes.
Collapse
Affiliation(s)
- Bipul R Acharya
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Jennifer S Fang
- Department of Molecular Biology & Biochemistry, University of California at Irvine, Irvine, CA, USA
| | - Erin D Jeffery
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nicholas W Chavkin
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Gael Genet
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Hema Vasavada
- Departments of Medicine and Genetics, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth A Nelson
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Gloria M Sheynkman
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Martin J Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Karen K Hirschi
- Department of Cell Biology, Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
- Departments of Medicine and Genetics, Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
Zeng H, Huang J, Ren J, Wang CK, Tang Z, Zhou H, Zhou Y, Shi H, Aditham A, Sui X, Chen H, Lo JA, Wang X. Spatially resolved single-cell translatomics at molecular resolution. Science 2023; 380:eadd3067. [PMID: 37384709 PMCID: PMC11146668 DOI: 10.1126/science.add3067] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 05/07/2023] [Indexed: 07/01/2023]
Abstract
The precise control of messenger RNA (mRNA) translation is a crucial step in posttranscriptional gene regulation of cellular physiology. However, it remains a challenge to systematically study mRNA translation at the transcriptomic scale with spatial and single-cell resolution. Here, we report the development of ribosome-bound mRNA mapping (RIBOmap), a highly multiplexed three-dimensional in situ profiling method to detect cellular translatome. RIBOmap profiling of 981 genes in HeLa cells revealed cell cycle-dependent translational control and colocalized translation of functional gene modules. We mapped 5413 genes in mouse brain tissues, yielding spatially resolved single-cell translatomic profiles for 119,173 cells and revealing cell type-specific and brain region-specific translational regulation, including translation remodeling during oligodendrocyte maturation. Our method detected widespread patterns of localized translation in neuronal and glial cells in intact brain tissue networks.
Collapse
Affiliation(s)
- Hu Zeng
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jiahao Huang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jingyi Ren
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Zefang Tang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Haowen Zhou
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yiming Zhou
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hailing Shi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Abhishek Aditham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Xin Sui
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Hongyu Chen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jennifer A. Lo
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xiao Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
22
|
Zhou M, Fang R, Colson L, Donovan KA, Hunkeler M, Song Y, Zhang C, Chen S, Lee DH, Bradshaw GA, Eisert R, Ye Y, Kalocsay M, Goldberg A, Fischer ES, Lu Y. HUWE1 Amplifies Ubiquitin Modifications to Broadly Stimulate Clearance of Proteins and Aggregates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542866. [PMID: 37398461 PMCID: PMC10312588 DOI: 10.1101/2023.05.30.542866] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Selective breakdown of proteins and aggregates is crucial for maintaining normal cellular activities and is involved in the pathogenesis of diverse diseases. How the cell recognizes and tags these targets in different structural states for degradation by the proteasome and autophagy pathways has not been well understood. Here, we discovered that a HECT-family ubiquitin ligase HUWE1 is broadly required for the efficient degradation of soluble factors and for the clearance of protein aggregates/condensates. Underlying this capacity of HUWE1 is a novel Ubiquitin-Directed ubiquitin Ligase (UDL) activity which recognizes both soluble substrates and aggregates that carry a high density of ubiquitin chains and rapidly expand the ubiquitin modifications on these targets. Ubiquitin signal amplification by HUWE1 recruits the ubiquitin-dependent segregase p97/VCP to process these targets for subsequent degradation or clearance. HUWE1 controls the cytotoxicity of protein aggregates, mediates Targeted Protein Degradation and regulates cell-cycle transitions with its UDL activity.
Collapse
|
23
|
Fischer L, Thievessen I. FUCCI Reporter Gene-Based Cell Cycle Analysis. Methods Mol Biol 2023; 2644:371-385. [PMID: 37142935 DOI: 10.1007/978-1-0716-3052-5_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
One of the most commonly assessed parameters in cellular analyses is the proliferative activity of a cell population. The fluorescence ubiquitin cell cycle indicator (FUCCI)-based system allows live and in vivo observation of cell cycle progression. Based on the mutually exclusive activity of two fluorescently labeled proteins cdt1 and geminin during the G0/1 and S/G2/M phases of the cell cycle, individual cells can be assigned to their respective cell cycle phase by fluorescence imaging of the nucleus. Here, we describe the generation of NIH/3T3 cells containing the FUCCI reporter system by lentiviral transduction and their use in 3D culture assays. The protocol can be adapted to other cell lines.
Collapse
Affiliation(s)
- Lena Fischer
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ingo Thievessen
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
24
|
Gupta RK, Mlcochova P. Cyclin D3 restricts SARS-CoV-2 envelope incorporation into virions and interferes with viral spread. EMBO J 2022; 41:e111653. [PMID: 36161661 PMCID: PMC9539236 DOI: 10.15252/embj.2022111653] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 01/13/2023] Open
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) presents a great threat to human health. The interplay between the virus and host plays a crucial role in successful virus replication and transmission. Understanding host-virus interactions are essential for the development of new COVID-19 treatment strategies. Here, we show that SARS-CoV-2 infection triggers redistribution of cyclin D1 and cyclin D3 from the nucleus to the cytoplasm, followed by proteasomal degradation. No changes to other cyclins or cyclin-dependent kinases were observed. Further, cyclin D depletion was independent of SARS-CoV-2-mediated cell cycle arrest in the early S phase or S/G2/M phase. Cyclin D3 knockdown by small-interfering RNA specifically enhanced progeny virus titres in supernatants. Finally, cyclin D3 co-immunoprecipitated with SARS-CoV-2 envelope (E) and membrane (M) proteins. We propose that cyclin D3 impairs the efficient incorporation of envelope protein into virions during assembly and is depleted during SARS-CoV-2 infection to restore efficient assembly and release of newly produced virions.
Collapse
Affiliation(s)
- Ravi K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID)CambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
- Africa Health Research InstituteDurbanSouth Africa
| | - Petra Mlcochova
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID)CambridgeUK
- Department of MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
25
|
Xu X, Duan S, Hua X, Li Z, He R, Zhang Z. Stable inheritance of H3.3-containing nucleosomes during mitotic cell divisions. Nat Commun 2022; 13:2514. [PMID: 35523900 PMCID: PMC9076889 DOI: 10.1038/s41467-022-30298-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 04/22/2022] [Indexed: 12/13/2022] Open
Abstract
Newly synthesized H3.1 and H3.3 histones are assembled into nucleosomes by different histone chaperones in replication-coupled and replication-independent pathways, respectively. However, it is not clear how parental H3.3 molecules are transferred following DNA replication, especially when compared to H3.1. Here, by monitoring parental H3.1- and H3.3-SNAP signals, we show that parental H3.3, like H3.1, are stably transferred into daughter cells. Moreover, Mcm2-Pola1 and Pole3-Pole4, two pathways involved in parental histone transfer based upon the analysis of modifications on parental histones, participate in the transfer of both H3.1 and H3.3 following DNA replication. Lastly, we found that Mcm2, Pole3 and Pole4 mutants defective in parental histone transfer show defects in chromosome segregation. These results indicate that in contrast to deposition of newly synthesized H3.1 and H3.3, transfer of parental H3.1 and H3.3 is mediated by these shared mechanisms, which contributes to epigenetic memory of gene expression and maintenance of genome stability.
Collapse
Affiliation(s)
- Xiaowei Xu
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Shoufu Duan
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Xu Hua
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Zhiming Li
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Richard He
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Zhiguo Zhang
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
26
|
KalantarMotamedi Y, Choi RJ, Koh SB, Bramhall JL, Fan TP, Bender A. Prediction and identification of synergistic compound combinations against pancreatic cancer cells. iScience 2021; 24:103080. [PMID: 34585118 PMCID: PMC8456050 DOI: 10.1016/j.isci.2021.103080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/28/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022] Open
Abstract
Resistance to current therapies is common for pancreatic cancer and hence novel treatment options are urgently needed. In this work, we developed and validated a computational method to select synergistic compound combinations based on transcriptomic profiles from both the disease and compound side, combined with a pathway scoring system, which was then validated prospectively by testing 30 compounds (and their combinations) on PANC-1 cells. Some compounds selected as single agents showed lower GI50 values than the standard of care, gemcitabine. Compounds suggested as combination agents with standard therapy gemcitabine based on the best performing scoring system showed on average 2.82-5.18 times higher synergies compared to compounds that were predicted to be active as single agents. Examples of highly synergistic in vitro validated compound pairs include gemcitabine combined with Entinostat, thioridazine, loperamide, scriptaid and Saracatinib. Hence, the computational approach presented here was able to identify synergistic compound combinations against pancreatic cancer cells.
Collapse
Affiliation(s)
- Yasaman KalantarMotamedi
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Ran Joo Choi
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Siang-Boon Koh
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Jo L. Bramhall
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Tai-Ping Fan
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
27
|
Molecular Changes Induced in Melanoma by Cell Culturing in 3D Alginate Hydrogels. Cancers (Basel) 2021; 13:cancers13164111. [PMID: 34439267 PMCID: PMC8394053 DOI: 10.3390/cancers13164111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary The research field of 3D cell cultivation in hydrogels is continuously growing. To be able to analyze the reaction of melanoma cells to 3D cultivation in alginate hydrogel on a molecular level, whole transcriptome sequencing was performed. Intriguingly, we could not only unravel differences between the gene regulation in 2D and 3D cultures but could also correlate the culture switch to the physiological process of tumor plasticity based on the observed patterns. Thereby, the role of EGR1 in controlling tumor plasticity and progression in melanoma was revealed. We conclude that the combination of cell culture models using biomaterials and whole transcriptome analysis leads to a deeper molecular understanding of cancer cells, herewith defining new therapeutic targets. Abstract Alginate hydrogels have been used as a biomaterial for 3D culturing for several years. Here, gene expression patterns in melanoma cells cultivated in 3D alginate are compared to 2D cultures. It is well-known that 2D cell culture is not resembling the complex in vivo situation well. However, the use of very intricate 3D models does not allow performing high-throughput screening and analysis is highly complex. 3D cell culture strategies in hydrogels will better mimic the in vivo situation while they maintain feasibility for large-scale analysis. As alginate is an easy-to-use material and due to its favorable properties, it is commonly applied as a bioink component in the growing field of cell encapsulation and biofabrication. Yet, only a little information about the transcriptome in 3D cultures in hydrogels like alginate is available. In this study, changes in the transcriptome based on RNA-Seq data by cultivating melanoma cells in 3D alginate are analyzed and reveal marked changes compared to cells cultured on usual 2D tissue culture plastic. Deregulated genes represent valuable cues to signaling pathways and molecules affected by the culture method. Using this as a model system for tumor cell plasticity and heterogeneity, EGR1 is determined to play an important role in melanoma progression.
Collapse
|
28
|
Koh SB, Ross K, Isakoff SJ, Melkonjan N, He L, Matissek KJ, Schultz A, Mayer EL, Traina TA, Carey LA, Rugo HS, Liu MC, Stearns V, Langenbucher A, Saladi SV, Ramaswamy S, Lawrence MS, Ellisen LW. RASAL2 Confers Collateral MEK/EGFR Dependency in Chemoresistant Triple-Negative Breast Cancer. Clin Cancer Res 2021; 27:4883-4897. [PMID: 34168046 DOI: 10.1158/1078-0432.ccr-21-0714] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/30/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE While chemotherapy remains the standard treatment for triple-negative breast cancer (TNBC), identifying and managing chemoresistant tumors has proven elusive. We sought to discover hallmarks and therapeutically actionable features of refractory TNBC through molecular analysis of primary chemoresistant TNBC specimens. EXPERIMENTAL DESIGN We performed transcriptional profiling of tumors from a phase II clinical trial of platinum chemotherapy for advanced TNBC (TBCRC-009), revealing a gene expression signature that identified de novo chemorefractory tumors. We then employed pharmacogenomic data mining, proteomic and other molecular studies to define the therapeutic vulnerabilities of these tumors. RESULTS We reveal the RAS-GTPase-activating protein (RAS-GAP) RASAL2 as an upregulated factor that mediates chemotherapy resistance but also an exquisite collateral sensitivity to combination MAP kinase kinase (MEK1/2) and EGFR inhibitors in TNBC. Mechanistically, RASAL2 GAP activity is required to confer kinase inhibitor sensitivity, as RASAL2-high TNBCs sustain basal RAS activity through suppression of negative feedback regulators SPRY1/2, together with EGFR upregulation. Consequently, RASAL2 expression results in failed feedback compensation upon co-inhibition of MEK1/2 and EGFR that induces synergistic apoptosis in vitro and in vivo. In patients with TNBC, high RASAL2 levels predict clinical chemotherapy response and long-term outcomes, and are associated via direct transcriptional regulation with activated oncogenic Yes-Associated Protein (YAP). Accordingly, chemorefractory patient-derived TNBC models exhibit YAP activation, high RASAL2 expression, and tumor regression in response to MEK/EGFR inhibitor combinations despite well-tolerated intermittent dosing. CONCLUSIONS These findings identify RASAL2 as a mediator of TNBC chemoresistance that rewires MAPK feedback and cross-talk to confer profound collateral sensitivity to combination MEK1/2 and EGFR inhibitors.
Collapse
Affiliation(s)
- Siang-Boon Koh
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Kenneth Ross
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts
| | - Steven J Isakoff
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Nsan Melkonjan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Lei He
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Karina J Matissek
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Andrew Schultz
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Erica L Mayer
- Harvard Medical School, Boston, Massachusetts.,Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Lisa A Carey
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hope S Rugo
- University of California San Francisco, San Francisco, California
| | - Minetta C Liu
- Georgetown Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Vered Stearns
- Johns Hopkins University and Sidney Kimmel Cancer Center, Baltimore, Maryland
| | - Adam Langenbucher
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Srinivas Vinod Saladi
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts.,Ludwig Center at Harvard, Harvard University, Boston, Massachusetts
| | - Michael S Lawrence
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts.,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts
| | - Leif W Ellisen
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts. .,Harvard Medical School, Boston, Massachusetts.,Ludwig Center at Harvard, Harvard University, Boston, Massachusetts
| |
Collapse
|
29
|
Slow-cycling (dormant) cancer cells in therapy resistance, cancer relapse and metastasis. Semin Cancer Biol 2021; 78:90-103. [PMID: 33979674 DOI: 10.1016/j.semcancer.2021.04.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
It is increasingly appreciated that cancer cell heterogeneity and plasticity constitute major barriers to effective clinical treatments and long-term therapeutic efficacy. Research in the past two decades suggest that virtually all treatment-naive human cancers harbor subsets of cancer cells that possess many of the cardinal features of normal stem cells. Such stem-like cancer cells, operationally defined as cancer stem cells (CSCs), are frequently quiescent and dynamically change and evolve during tumor progression and therapeutic interventions. Intrinsic tumor cell heterogeneity is reflected in a different aspect in that tumors also harbor a population of slow-cycling cells (SCCs) that are not in the proliferative cell cycle and thus are intrinsically refractory to anti-mitotic drugs. In this Perspective, we focus our discussions on SCCs in cancer and on various methodologies that can be employed to enrich and purify SCCs, compare the similarities and differences between SCCs, CSCs and cancer cells undergoing EMT, and present evidence for the involvement of SCCs in surviving anti-neoplastic treatments, mediating tumor relapse, maintaining tumor dormancy and mediating metastatic dissemination. Our discussions make it clear that an in-depth understanding of the biological properties of SCCs in cancer will be instrumental to developing new therapeutic strategies to prevent tumor relapse and distant metastasis.
Collapse
|
30
|
Ghannoum S, Antos K, Leoncio Netto W, Gomes C, Köhn-Luque A, Farhan H. CellMAPtracer: A User-Friendly Tracking Tool for Long-Term Migratory and Proliferating Cells Associated with FUCCI Systems. Cells 2021; 10:cells10020469. [PMID: 33671785 PMCID: PMC7927118 DOI: 10.3390/cells10020469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/06/2021] [Accepted: 02/18/2021] [Indexed: 01/23/2023] Open
Abstract
Cell migration is a fundamental biological process of key importance in health and disease. Advances in imaging techniques have paved the way to monitor cell motility. An ever-growing collection of computational tools to track cells has improved our ability to analyze moving cells. One renowned goal in the field is to provide tools that track cell movement as comprehensively and automatically as possible. However, fully automated tracking over long intervals of time is challenged by dividing cells, thus calling for a combination of automated and supervised tracking. Furthermore, after the emergence of various experimental tools to monitor cell-cycle phases, it is of relevance to integrate the monitoring of cell-cycle phases and motility. We developed CellMAPtracer, a multiplatform tracking system that achieves that goal. It can be operated as a conventional, automated tracking tool of single cells in numerous imaging applications. However, CellMAPtracer also allows adjusting tracked cells in a semiautomated supervised fashion, thereby improving the accuracy and facilitating the long-term tracking of migratory and dividing cells. CellMAPtracer is available with a user-friendly graphical interface and does not require any coding or programming skills. CellMAPtracer is compatible with two- and three-color fluorescent ubiquitination-based cell-cycle indicator (FUCCI) systems and allows the user to accurately monitor various migration parameters throughout the cell cycle, thus having great potential to facilitate new discoveries in cell biology.
Collapse
Affiliation(s)
- Salim Ghannoum
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway;
- Correspondence: (S.G.); (K.A.); Tel.: +46-76-577-0129 (S.G.)
| | - Kamil Antos
- Department of Integrative Medical Biology, Umeå University, 90736 Umeå, Sweden
- Correspondence: (S.G.); (K.A.); Tel.: +46-76-577-0129 (S.G.)
| | - Waldir Leoncio Netto
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (W.L.N.); (A.K.-L.)
| | - Cecil Gomes
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA;
| | - Alvaro Köhn-Luque
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (W.L.N.); (A.K.-L.)
| | - Hesso Farhan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway;
- Institute of Pathophysiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
31
|
Lomakin AJ, Cattin CJ, Cuvelier D, Alraies Z, Molina M, Nader GPF, Srivastava N, Sáez PJ, Garcia-Arcos JM, Zhitnyak IY, Bhargava A, Driscoll MK, Welf ES, Fiolka R, Petrie RJ, De Silva NS, González-Granado JM, Manel N, Lennon-Duménil AM, Müller DJ, Piel M. The nucleus acts as a ruler tailoring cell responses to spatial constraints. Science 2020; 370:eaba2894. [PMID: 33060332 PMCID: PMC8059074 DOI: 10.1126/science.aba2894] [Citation(s) in RCA: 326] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 06/29/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
The microscopic environment inside a metazoan organism is highly crowded. Whether individual cells can tailor their behavior to the limited space remains unclear. In this study, we found that cells measure the degree of spatial confinement by using their largest and stiffest organelle, the nucleus. Cell confinement below a resting nucleus size deforms the nucleus, which expands and stretches its envelope. This activates signaling to the actomyosin cortex via nuclear envelope stretch-sensitive proteins, up-regulating cell contractility. We established that the tailored contractile response constitutes a nuclear ruler-based signaling pathway involved in migratory cell behaviors. Cells rely on the nuclear ruler to modulate the motive force that enables their passage through restrictive pores in complex three-dimensional environments, a process relevant to cancer cell invasion, immune responses, and embryonic development.
Collapse
Affiliation(s)
- A J Lomakin
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences (ÖAW), Vienna, Austria
- Medical University of Vienna (MUV), Vienna, Austria
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London, UK
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - C J Cattin
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - D Cuvelier
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - Z Alraies
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - M Molina
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, King's College London, London, UK
| | - G P F Nader
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - N Srivastava
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - P J Sáez
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - J M Garcia-Arcos
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| | - I Y Zhitnyak
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
- N.N. Blokhin Medical Research Center of Oncology, Moscow, Russia
| | - A Bhargava
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - M K Driscoll
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - E S Welf
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R Fiolka
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R J Petrie
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - N S De Silva
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | - J M González-Granado
- LamImSys Lab, Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid, Spain
| | - N Manel
- Institut Curie, PSL Research University, INSERM, U 932, Paris, France
| | | | - D J Müller
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
| | - M Piel
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France.
- Institut Pierre Gilles de Gennes, PSL Research University, Paris, France
| |
Collapse
|
32
|
Metz EP, Wuebben EL, Wilder PJ, Cox JL, Datta K, Coulter D, Rizzino A. Tumor quiescence: elevating SOX2 in diverse tumor cell types downregulates a broad spectrum of the cell cycle machinery and inhibits tumor growth. BMC Cancer 2020; 20:941. [PMID: 32998722 PMCID: PMC7528478 DOI: 10.1186/s12885-020-07370-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/31/2020] [Indexed: 11/17/2022] Open
Abstract
Background Quiescent tumor cells pose a major clinical challenge due to their ability to resist conventional chemotherapies and to drive tumor recurrence. Understanding the molecular mechanisms that promote quiescence of tumor cells could help identify therapies to eliminate these cells. Significantly, recent studies have determined that the function of SOX2 in cancer cells is highly dose dependent. Specifically, SOX2 levels in tumor cells are optimized to promote tumor growth: knocking down or elevating SOX2 inhibits proliferation. Furthermore, recent studies have shown that quiescent tumor cells express higher levels of SOX2 compared to adjacent proliferating cells. Currently, the mechanisms through which elevated levels of SOX2 restrict tumor cell proliferation have not been characterized. Methods To understand how elevated levels of SOX2 restrict the proliferation of tumor cells, we engineered diverse types of tumor cells for inducible overexpression of SOX2. Using these cells, we examined the effects of elevating SOX2 on their proliferation, both in vitro and in vivo. In addition, we examined how elevating SOX2 influences their expression of cyclins, cyclin-dependent kinases (CDKs), and p27Kip1. Results Elevating SOX2 in diverse tumor cell types led to growth inhibition in vitro. Significantly, elevating SOX2 in vivo in pancreatic ductal adenocarcinoma, medulloblastoma, and prostate cancer cells induced a reversible state of tumor growth arrest. In all three tumor types, elevation of SOX2 in vivo quickly halted tumor growth. Remarkably, tumor growth resumed rapidly when SOX2 returned to endogenous levels. We also determined that elevation of SOX2 in six tumor cell lines decreased the levels of cyclins and CDKs that control each phase of the cell cycle, while upregulating p27Kip1. Conclusions Our findings indicate that elevating SOX2 above endogenous levels in a diverse set of tumor cell types leads to growth inhibition both in vitro and in vivo. Moreover, our findings indicate that SOX2 can function as a master regulator by controlling the expression of a broad spectrum of cell cycle machinery. Importantly, our SOX2-inducible tumor studies provide a novel model system for investigating the molecular mechanisms by which elevated levels of SOX2 restrict cell proliferation and tumor growth.
Collapse
Affiliation(s)
- Ethan P Metz
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Erin L Wuebben
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Phillip J Wilder
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center Fred & Pamela Buffett Cancer Center, Omaha, NE, 68198-6805, USA
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Donald Coulter
- Department of Pediatrics, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Angie Rizzino
- Eppley Institute for Research in Cancer and Allied Diseases Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center Fred & Pamela Buffett Cancer Center, Omaha, NE, 68198-6805, USA. .,Department of Biochemistry and Molecular Biology Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
| |
Collapse
|
33
|
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
|
34
|
Sweeney MA, Iakova P, Maneix L, Shih FY, Cho HE, Sahin E, Catic A. The ubiquitin ligase Cullin-1 associates with chromatin and regulates transcription of specific c-MYC target genes. Sci Rep 2020; 10:13942. [PMID: 32811853 PMCID: PMC7435197 DOI: 10.1038/s41598-020-70610-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022] Open
Abstract
Transcription is regulated through a dynamic interplay of DNA-associated proteins, and the composition of gene-regulatory complexes is subject to continuous adjustments. Protein alterations include post-translational modifications and elimination of individual polypeptides. Spatially and temporally controlled protein removal is, therefore, essential for gene regulation and accounts for the short half-life of many transcription factors. The ubiquitin-proteasome system is responsible for site- and target-specific ubiquitination and protein degradation. Specificity of ubiquitination is conferred by ubiquitin ligases. Cullin-RING complexes, the largest family of ligases, require multi-unit assembly around one of seven cullin proteins. To investigate the direct role of cullins in ubiquitination of DNA-bound proteins and in gene regulation, we analyzed their subcellular locations and DNA-affinities. We found CUL4A and CUL7 to be largely excluded from the nucleus, whereas CUL4B was primarily nuclear. CUL1,2,3, and 5 showed mixed cytosolic and nuclear expression. When analyzing chromatin affinity of individual cullins, we discovered that CUL1 preferentially associated with active promoter sequences and co-localized with 23% of all DNA-associated protein degradation sites. CUL1 co-distributed with c-MYC and specifically repressed nuclear-encoded mitochondrial and splicing-associated genes. These studies underscore the relevance of spatial control in chromatin-associated protein ubiquitination and define a novel role for CUL1 in gene repression.
Collapse
Affiliation(s)
- Melanie A Sweeney
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Polina Iakova
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Laure Maneix
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Fu-Yuan Shih
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Hannah E Cho
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Rice University Undergraduate School of Social Sciences, Houston, TX, USA
| | - Ergun Sahin
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Andre Catic
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
35
|
Ghram M, Bonnet-Magnaval F, Hotea DI, Doran B, Ly S, DesGroseillers L. Staufen1 is Essential for Cell-Cycle Transitions and Cell Proliferation Via the Control of E2F1 Expression. J Mol Biol 2020; 432:3881-3897. [DOI: 10.1016/j.jmb.2020.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/16/2022]
|
36
|
Andrei L, Kasas S, Ochoa Garrido I, Stanković T, Suárez Korsnes M, Vaclavikova R, Assaraf YG, Pešić M. Advanced technological tools to study multidrug resistance in cancer. Drug Resist Updat 2020; 48:100658. [DOI: 10.1016/j.drup.2019.100658] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
|
37
|
Albrengues J, Shields MA, Ng D, Park CG, Ambrico A, Poindexter ME, Upadhyay P, Uyeminami DL, Pommier A, Küttner V, Bružas E, Maiorino L, Bautista C, Carmona EM, Gimotty PA, Fearon DT, Chang K, Lyons SK, Pinkerton KE, Trotman LC, Goldberg MS, Yeh JTH, Egeblad M. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018; 361:eaao4227. [PMID: 30262472 PMCID: PMC6777850 DOI: 10.1126/science.aao4227] [Citation(s) in RCA: 993] [Impact Index Per Article: 141.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/03/2018] [Indexed: 12/11/2022]
Abstract
Cancer cells from a primary tumor can disseminate to other tissues, remaining dormant and clinically undetectable for many years. Little is known about the cues that cause these dormant cells to awaken, resume proliferating, and develop into metastases. Studying mouse models, we found that sustained lung inflammation caused by tobacco smoke exposure or nasal instillation of lipopolysaccharide converted disseminated, dormant cancer cells to aggressively growing metastases. Sustained inflammation induced the formation of neutrophil extracellular traps (NETs), and these were required for awakening dormant cancer. Mechanistic analysis revealed that two NET-associated proteases, neutrophil elastase and matrix metalloproteinase 9, sequentially cleaved laminin. The proteolytically remodeled laminin induced proliferation of dormant cancer cells by activating integrin α3β1 signaling. Antibodies against NET-remodeled laminin prevented awakening of dormant cells. Therapies aimed at preventing dormant cell awakening could potentially prolong the survival of cancer patients.
Collapse
Affiliation(s)
- Jean Albrengues
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Mario A Shields
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - David Ng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Chun Gwon Park
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02215, USA
| | | | - Morgan E Poindexter
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616, USA
| | - Priya Upadhyay
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616, USA
| | - Dale L Uyeminami
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616, USA
| | - Arnaud Pommier
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Victoria Küttner
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Emilis Bružas
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Watson School of Biological Sciences, Cold Spring Harbor, NY 11724, USA
| | - Laura Maiorino
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Watson School of Biological Sciences, Cold Spring Harbor, NY 11724, USA
| | | | - Ellese M Carmona
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02215, USA
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, UK
- Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Scott K Lyons
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616, USA
| | - Lloyd C Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Michael S Goldberg
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02215, USA
| | - Johannes T-H Yeh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
38
|
Koh SB, Wallez Y, Dunlop CR, Bernaldo de Quirós Fernández S, Bapiro TE, Richards FM, Jodrell DI. Mechanistic Distinctions between CHK1 and WEE1 Inhibition Guide the Scheduling of Triple Therapy with Gemcitabine. Cancer Res 2018; 78:3054-3066. [PMID: 29735549 PMCID: PMC5985963 DOI: 10.1158/0008-5472.can-17-3932] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/14/2018] [Accepted: 04/04/2018] [Indexed: 12/31/2022]
Abstract
Combination of cytotoxic therapy with emerging DNA damage response inhibitors (DDRi) has been limited by tolerability issues. However, the goal of most combination trials has been to administer DDRi with standard-of-care doses of chemotherapy. We hypothesized that mechanism-guided treatment scheduling could reduce the incidence of dose-limiting toxicities and enable tolerable multitherapeutic regimens. Integrative analyses of mathematical modeling and single-cell assays distinguished the synergy kinetics of WEE1 inhibitor (WEE1i) from CHEK1 inhibitor (CHK1i) by potency, spatiotemporal perturbation, and mitotic effects when combined with gemcitabine. These divergent properties collectively supported a triple-agent strategy, whereby a pulse of gemcitabine and CHK1i followed by WEE1i durably suppressed tumor cell growth. In xenografts, CHK1i exaggerated replication stress without mitotic CDK hyperactivation, enriching a geminin-positive subpopulation and intratumoral gemcitabine metabolite. Without overt toxicity, addition of WEE1i to low-dose gemcitabine and CHK1i was most effective in tumor control compared with single and double agents. Overall, our work provides quantitative insights into the mechanisms of DDRi chemosensitization, leading to the rational development of a tolerable multitherapeutic regimen.Significance: Multiple lines of mechanistic insight regarding DNA damage response inhibitors rationally guide the preclinical development of a tolerable multitherapeutic regimen.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/11/3054/F1.large.jpg Cancer Res; 78(11); 3054-66. ©2018 AACR.
Collapse
Affiliation(s)
- Siang-Boon Koh
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom.
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Yann Wallez
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Charles R Dunlop
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Tashinga E Bapiro
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Frances M Richards
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Duncan I Jodrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
39
|
Mittal R, Nguyen D, Patel AP, Debs LH, Mittal J, Yan D, Eshraghi AA, Van De Water TR, Liu XZ. Recent Advancements in the Regeneration of Auditory Hair Cells and Hearing Restoration. Front Mol Neurosci 2017; 10:236. [PMID: 28824370 PMCID: PMC5534485 DOI: 10.3389/fnmol.2017.00236] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/11/2017] [Indexed: 12/18/2022] Open
Abstract
Neurosensory responses of hearing and balance are mediated by receptors in specialized neuroepithelial sensory cells. Any disruption of the biochemical and molecular pathways that facilitate these responses can result in severe deficits, including hearing loss and vestibular dysfunction. Hearing is affected by both environmental and genetic factors, with impairment of auditory function being the most common neurosensory disorder affecting 1 in 500 newborns, as well as having an impact on the majority of elderly population. Damage to auditory sensory cells is not reversible, and if sufficient damage and cell death have taken place, the resultant deficit may lead to permanent deafness. Cochlear implants are considered to be one of the most successful and consistent treatments for deaf patients, but only offer limited recovery at the expense of loss of residual hearing. Recently there has been an increased interest in the auditory research community to explore the regeneration of mammalian auditory hair cells and restoration of their function. In this review article, we examine a variety of recent therapies, including genetic, stem cell and molecular therapies as well as discussing progress being made in genome editing strategies as applied to the restoration of hearing function.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Desiree Nguyen
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Amit P. Patel
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Luca H. Debs
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Adrien A. Eshraghi
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Thomas R. Van De Water
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
| | - Xue Z. Liu
- Department of Otolaryngology, University of Miami Miller School of MedicineMiami, FL, United States
- Department of Otolaryngology, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
40
|
Grah JS, Harrington JA, Koh SB, Pike JA, Schreiner A, Burger M, Schönlieb CB, Reichelt S. Mathematical imaging methods for mitosis analysis in live-cell phase contrast microscopy. Methods 2017; 115:91-99. [PMID: 28189773 PMCID: PMC6414815 DOI: 10.1016/j.ymeth.2017.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 02/04/2017] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
In this paper we propose a workflow to detect and track mitotic cells in time-lapse microscopy image sequences. In order to avoid the requirement for cell lines expressing fluorescent markers and the associated phototoxicity, phase contrast microscopy is often preferred over fluorescence microscopy in live-cell imaging. However, common specific image characteristics complicate image processing and impede use of standard methods. Nevertheless, automated analysis is desirable due to manual analysis being subjective, biased and extremely time-consuming for large data sets. Here, we present the following workflow based on mathematical imaging methods. In the first step, mitosis detection is performed by means of the circular Hough transform. The obtained circular contour subsequently serves as an initialisation for the tracking algorithm based on variational methods. It is sub-divided into two parts: in order to determine the beginning of the whole mitosis cycle, a backwards tracking procedure is performed. After that, the cell is tracked forwards in time until the end of mitosis. As a result, the average of mitosis duration and ratios of different cell fates (cell death, no division, division into two or more daughter cells) can be measured and statistics on cell morphologies can be obtained. All of the tools are featured in the user-friendly MATLAB®Graphical User Interface MitosisAnalyser.
Collapse
Affiliation(s)
- Joana Sarah Grah
- University of Cambridge, Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, United Kingdom.
| | - Jennifer Alison Harrington
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Siang Boon Koh
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Jeremy Andrew Pike
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Alexander Schreiner
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| | - Martin Burger
- Westfälische Wilhelms-Universität Münster, Institute for Computational and Applied Mathematics, Einsteinstrasse 62, 48149 Münster, Germany
| | - Carola-Bibiane Schönlieb
- University of Cambridge, Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, United Kingdom
| | - Stefanie Reichelt
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|