1
|
Crncec A, Lau HW, Ng LY, Ma HT, Mak JP, Choi HF, Yeung TK, Poon RYC. Plasticity of mitotic cyclins in promoting the G2-M transition. J Cell Biol 2025; 224:e202409219. [PMID: 40202486 PMCID: PMC11980681 DOI: 10.1083/jcb.202409219] [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: 09/30/2024] [Revised: 01/31/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
Cyclins and cyclin-dependent kinases (CDKs) orchestrate key events in the cell cycle. However, the uniqueness of individual mitotic cyclins has been a long-standing puzzle. By rapidly removing cyclins in G2 human cells, we found that deficiency of B-type cyclins attenuates mitotic onset and uncouples the G2-M kinase network from mitosis, resulting in sustained activation of PLK1 and cyclin A-CDK1. This culminates in mitotic slippage without completing nuclear envelope breakdown. Remarkably, elevating cyclin A several-fold above its endogenous level is adequate to restore mitosis, allowing cells to survive without B-type cyclins. In contrast, cyclin A is rate-limiting but not essential for G2-M due to compensation by endogenous cyclin B1-CDK2, a non-canonical pair. These findings challenge the traditional indispensable roles of different cyclins and highlight their plasticity. Due to the high malleability of the A- and B-type cyclins, cancer cells may be able to place different weights on different cyclins, while maintaining sufficient CDK activities for successful mitosis.
Collapse
Affiliation(s)
- Adrijana Crncec
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ho Wai Lau
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Lau Yan Ng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hoi Tang Ma
- Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Joyce P.Y. Mak
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hon Fung Choi
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Tsz Kwan Yeung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Yat Choi Poon
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
2
|
Gong Y, Dong M, Feng X, Zhang N, Cui X, Wang L, Qi Q, Kuok CF, Jiang Q, Bi S. CS-6-induced p62 accumulation exacerbates DNA damage in colorectal cancer. Front Pharmacol 2025; 16:1568339. [PMID: 40417224 PMCID: PMC12098580 DOI: 10.3389/fphar.2025.1568339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025] Open
Abstract
Background Gamabufotalin (CS-6), a bufadienolide derived from Chansu, has been reported to exhibit anti-tumor effects in various cancers, including glioblastoma, nonsmall cell lung cancer, and breast cancer. However,its role in colorectal cancer (CRC) remains unexplored. Objective Our study aimed to evaluate the inhibition of CS-6 to CRC cells by cell viability assay, colony formation assay, comet assay, and cell cycle analysis firstly. And its molecular mechanism was studied by immunofluorescence (IF) assay, western blot (WB) assay, siRNA transfection, protein-protein interaction (PPI) network and co-immunoprecipitation (Co-IP) assay. Finally, the in vivo antitumor assessments of CS-6 on colorectal cancer was validated through an transplant colorectal cancer model. Results CS-6 treatment significantly inhibited CRC SW620 and DLD1 cell viability and colony formation in vitro. Furthermore, CS-6 treatment-induced DNA damage and cell cycle arrest in SW620 and DLD1 cells. The western blot assay revealed that CS-6 treatment upregulated p62 expression. Knockdown of p62 in this study significantly alleviated CS-6-induced DNA damage and the downregulation of cyclin expression in SW620 and DLD1 cells. Additionally, the results indicated increased expression of microtubuleassociated protein I/II light chain 3II (LC3II) and reduced binding between B-cell lymphoma-2 (Bcl2) and beclin-1, suggesting that CS-6 treatment activated early-stage autophagy in CRC cells. However, inhibition of latestage autophagy and autophagy-related protein 5 (ATG5) with chloroquine and si-ATG5, respectively, further indicated that CS-6-induced autophagy defects led to p62 accumulation, exacerbated cell proliferation inhibition, and aggravated DNA damage. Intraperitoneal injection with CS-6 inhibited tumor growth in nude mice with colorectal cancer, and promoted the protein expression of phosphorylated H2A histone family member X (γH2AX), p62, phosphorylated Ataxia-telangiectasia mutated kinase (p-ATM) and LC3 I/II. Conclusion This study suggests that CS-6 may exert its anti-tumor effects in CRC by inducing autophagy defects, resulting in p62 accumulation and DNA damage in vitro and in vivo.
Collapse
Affiliation(s)
- Yitong Gong
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
- School of Humanities, Ludong University, Yantai, Shandong, China
| | - Menghan Dong
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaofei Feng
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
- Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Naiyu Zhang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Xuehui Cui
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Liushuyue Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Qi Qi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Chiu-fai Kuok
- Faculty of Health Sciences and Sports, Macao Polytechnic University, Macao, SAR China
| | - Qingling Jiang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Sixue Bi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
3
|
Gobran M, Politi AZ, Welp L, Jakobi J, Urlaub H, Lenart P. PLK1 inhibition delays mitotic entry revealing changes to the phosphoproteome of mammalian cells early in division. EMBO J 2025; 44:1891-1920. [PMID: 40033019 PMCID: PMC11962124 DOI: 10.1038/s44318-025-00400-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Polo-like kinase 1 (PLK1) is a conserved regulator of cell division. During mitotic prophase, PLK1 contributes to the activation of the cyclin-dependent kinase 1 (CDK1). However, the exact functions of PLK1 in prophase remain incompletely understood. Here, we show that PLK1 inhibition in synchronous G2 cell populations of multiple mammalian cell lines delays or prevents mitotic entry with high variability between individual cells. Using a mathematical model, we recapitulate this phenomenon and provide an explanation for the observed phenotypic variability. We show that PLK1-inhibited cells are delayed in a prophase-like state with low CDK1 activity that increases slowly and gradually over hours. These cells display progressively condensing chromosomes, increased microtubule dynamics, and reorganization of the actin cortex, while the nuclear envelope remains intact. We characterize this state further by phosphoproteomics, revealing phosphorylation of regulators of chromatin organization and the cytoskeleton consistent with the cellular phenotypes. Together, our results indicate that PLK1 inhibition stabilizes cells in a prophase-like state with low CDK1 activity displaying a specific set of early mitotic phosphorylation events.
Collapse
Affiliation(s)
- Monica Gobran
- Research Group Cytoskeletal Dynamics in Oocytes, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany
| | - Antonio Z Politi
- Research Group Cytoskeletal Dynamics in Oocytes, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany
| | - Luisa Welp
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany
- Bioanalytics, Institute of Clinical Chemistry, University Medical Center Göttingen, 40 Robert Koch Strasse, 37075, Göttingen, Germany
| | - Jasmin Jakobi
- Research Group Cytoskeletal Dynamics in Oocytes, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany
- Bioanalytics, Institute of Clinical Chemistry, University Medical Center Göttingen, 40 Robert Koch Strasse, 37075, Göttingen, Germany
| | - Peter Lenart
- Research Group Cytoskeletal Dynamics in Oocytes, Max Planck Institute for Multidisciplinary Sciences, 11 Am Fassberg, 37077, Göttingen, Germany.
| |
Collapse
|
4
|
Prifti DK, Lauzier A, Garand C, Calvo E, Devillers R, Roy S, Dos Santos A, Descombes L, Trudel B, Laplante M, Bordeleau F, Elowe S. ARHGEF17/TEM4 regulates the cell cycle through control of G1 progression. J Cell Biol 2025; 224:e202311194. [PMID: 39903211 PMCID: PMC11792891 DOI: 10.1083/jcb.202311194] [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: 04/23/2024] [Revised: 10/17/2024] [Accepted: 12/10/2024] [Indexed: 02/06/2025] Open
Abstract
The Ras homolog (Rho) small GTPases coordinate diverse cellular functions including cell morphology, adhesion and motility, cell cycle progression, survival, and apoptosis via their role in regulating the actin cytoskeleton. The upstream regulators for many of these functions are unknown. ARHGEF17 (also known as TEM4) is a Rho family guanine nucleotide exchange factor (GEF) implicated in cell migration, cell-cell junction formation, and the mitotic checkpoint. In this study, we characterize the regulation of the cell cycle by TEM4. We demonstrate that TEM4-depleted cells exhibit multiple defects in mitotic entry and duration, spindle morphology, and spindle orientation. In addition, TEM4 insufficiency leads to excessive cortical actin polymerization and cell rounding defects. Mechanistically, we demonstrate that TEM4-depleted cells delay in G1 as a consequence of decreased expression of the proproliferative transcriptional co-activator YAP. TEM4-depleted cells that progress through to mitosis do so with decreased levels of cyclin B as a result of attenuated expression of CCNB1. Importantly, cyclin B overexpression in TEM4-depleted cells largely rescues mitotic progression and chromosome segregation defects in anaphase. Our study thus illustrates the consequences of Rho signaling imbalance on cell cycle progression and identifies TEM4 as the first GEF governing Rho GTPase-mediated regulation of G1/S.
Collapse
Affiliation(s)
- Diogjena Katerina Prifti
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Annie Lauzier
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Chantal Garand
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Eva Calvo
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Romain Devillers
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
| | - Suparba Roy
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Alexsandro Dos Santos
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Laurence Descombes
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| | - Benjamin Trudel
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval (LOEX), Québec, Canada
| | - Mathieu Laplante
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de pneumologie de Québec (CRIUCPQ), Faculté de Médecine, Université Laval, Québec, Canada
| | - François Bordeleau
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Cancer, Québec, Canada
- Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec City, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval (LOEX), Québec, Canada
| | - Sabine Elowe
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe de Réproduction, Santé de la Mère et de l’Enfant, Québec, Canada
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l’Ingénierie et les Applications des protéines, Québec, Canada
- Département de Pédiatrie, Faculté de Médicine, Université Laval, Québec, Canada
- Centre de Recherche sur le Cancer de l’Université Laval, Québec, Canada
| |
Collapse
|
5
|
Cohen BE. The Role of the Swollen State in Cell Proliferation. J Membr Biol 2025; 258:1-13. [PMID: 39482485 DOI: 10.1007/s00232-024-00328-x] [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: 08/12/2024] [Accepted: 10/17/2024] [Indexed: 11/03/2024]
Abstract
Cell swelling is known to be involved in various stages of the growth of plant cells and microorganisms but in mammalian cells how crucial a swollen state is for determining the fate of the cellular proliferation remains unclear. Recent evidence has increased our understanding of how the loss of the cell surface interactions with the extracellular matrix at early mitosis decreases the membrane tension triggering curvature changes in the plasma membrane and the activation of the sodium/hydrogen (Na +/H +) exchanger (NHE1) that drives osmotic swelling. Such a swollen state is temporary, but it is critical to alter essential membrane biophysical parameters that are required to activate Ca2 + channels and modulate the opening of K + channels involved in setting the membrane potential. A decreased membrane potential across the mitotic cell membrane enhances the clustering of Ras proteins involved in the Ca2 + and cytoskeleton-driven events that lead to cell rounding. Changes in the external mechanical and osmotic forces also have an impact on the lipid composition of the plasma membrane during mitosis.
Collapse
|
6
|
Yu K, Wang GM, Guo SS, Bassermann F, Fässler R. The USP12/46 deubiquitinases protect integrins from ESCRT-mediated lysosomal degradation. EMBO Rep 2024; 25:5687-5718. [PMID: 39506038 PMCID: PMC11624278 DOI: 10.1038/s44319-024-00300-9] [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: 05/14/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
The functions of integrins are tightly regulated via multiple mechanisms including trafficking and degradation. Integrins are repeatedly internalized, routed into the endosomal system and either degraded by the lysosome or recycled back to the plasma membrane. The ubiquitin system dictates whether internalized proteins are degraded or recycled. Here, we use a genetic screen and proximity-dependent biotin identification to identify deubiquitinase(s) that control integrin surface levels. We find that a ternary deubiquitinating complex, comprised of USP12 (or the homologous USP46), WDR48 and WDR20, stabilizes β1 integrin (Itgb1) by preventing ESCRT-mediated lysosomal degradation. Mechanistically, the USP12/46-WDR48-WDR20 complex removes ubiquitin from the cytoplasmic tail of internalized Itgb1 in early endosomes, which in turn prevents ESCRT-mediated sorting and Itgb1 degradation.
Collapse
Affiliation(s)
- Kaikai Yu
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Guan M Wang
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Shiny Shengzhen Guo
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Florian Bassermann
- Department of Medicine III, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Heidelberg, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
7
|
Kors S, Schlaitz AL. Dynamic remodelling of the endoplasmic reticulum for mitosis. J Cell Sci 2024; 137:jcs261444. [PMID: 39584405 DOI: 10.1242/jcs.261444] [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: 11/26/2024] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic and continuous membrane network with roles in many cellular processes. The importance and maintenance of ER structure and function have been extensively studied in interphase cells, yet recent findings also indicate crucial roles of the ER in mitosis. During mitosis, the ER is remodelled significantly with respect to composition and morphology but persists as a continuous network. The ER interacts with microtubules, actin and intermediate filaments, and concomitant with the mitotic restructuring of all cytoskeletal systems, ER dynamics and distribution change. The ER is a metabolic hub and several examples of altered ER functions during mitosis have been described. However, we lack an overall understanding of the ER metabolic pathways and functions that are active during mitosis. In this Review, we will discuss mitotic changes to the ER at different organizational levels to explore how the mitotic ER, with its distinct properties, might support cell division.
Collapse
Affiliation(s)
- Suzan Kors
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| | - Anne-Lore Schlaitz
- Heidelberg University Biochemistry Center (BZH), 69120 Heidelberg, Germany
| |
Collapse
|
8
|
Liao Y, Zhang W, Liu Y, Zhu C, Zou Z. The role of ubiquitination in health and disease. MedComm (Beijing) 2024; 5:e736. [PMID: 39329019 PMCID: PMC11424685 DOI: 10.1002/mco2.736] [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: 02/11/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Ubiquitination is an enzymatic process characterized by the covalent attachment of ubiquitin to target proteins, thereby modulating their degradation, transportation, and signal transduction. By precisely regulating protein quality and quantity, ubiquitination is essential for maintaining protein homeostasis, DNA repair, cell cycle regulation, and immune responses. Nevertheless, the diversity of ubiquitin enzymes and their extensive involvement in numerous biological processes contribute to the complexity and variety of diseases resulting from their dysregulation. The ubiquitination process relies on a sophisticated enzymatic system, ubiquitin domains, and ubiquitin receptors, which collectively impart versatility to the ubiquitination pathway. The widespread presence of ubiquitin highlights its potential to induce pathological conditions. Ubiquitinated proteins are predominantly degraded through the proteasomal system, which also plays a key role in regulating protein localization and transport, as well as involvement in inflammatory pathways. This review systematically delineates the roles of ubiquitination in maintaining protein homeostasis, DNA repair, genomic stability, cell cycle regulation, cellular proliferation, and immune and inflammatory responses. Furthermore, the mechanisms by which ubiquitination is implicated in various pathologies, alongside current modulators of ubiquitination are discussed. Enhancing our comprehension of ubiquitination aims to provide novel insights into diseases involving ubiquitination and to propose innovative therapeutic strategies for clinical conditions.
Collapse
Affiliation(s)
- Yan Liao
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Wangzheqi Zhang
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Yang Liu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Chenglong Zhu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Zui Zou
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| |
Collapse
|
9
|
He J, Zeng X, Wang C, Wang E, Li Y. Antibody-drug conjugates in cancer therapy: mechanisms and clinical studies. MedComm (Beijing) 2024; 5:e671. [PMID: 39070179 PMCID: PMC11283588 DOI: 10.1002/mco2.671] [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: 01/05/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Antibody-drug conjugates (ADCs) consist of monoclonal antibodies that target tumor cells and cytotoxic drugs linked through linkers. By leveraging antibodies' targeting properties, ADCs deliver cytotoxic drugs into tumor cells via endocytosis after identifying the tumor antigen. This precise method aims to kill tumor cells selectively while minimizing harm to normal cells, offering safe and effective therapeutic benefits. Recent years have seen significant progress in antitumor treatment with ADC development, providing patients with new and potent treatment options. With over 300 ADCs explored for various tumor indications and some already approved for clinical use, challenges such as resistance due to factors like antigen expression, ADC processing, and payload have emerged. This review aims to outline the history of ADC development, their structure, mechanism of action, recent composition advancements, target selection, completed and ongoing clinical trials, resistance mechanisms, and intervention strategies. Additionally, it will delve into the potential of ADCs with novel markers, linkers, payloads, and innovative action mechanisms to enhance cancer treatment options. The evolution of ADCs has also led to the emergence of combination therapy as a new therapeutic approach to improve drug efficacy.
Collapse
Affiliation(s)
- Jun He
- Department of General Surgery Jiande Branch of the Second Affiliated Hospital, School of Medicine, Zhejiang University Jiande Zhejiang China
| | - Xianghua Zeng
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Chunmei Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Enwen Wang
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| | - Yongsheng Li
- Department of Medical Oncology Chongqing University Cancer Hospital Chongqing China
| |
Collapse
|
10
|
Yang C, Zhang W, Xiang S, Chen L, Chun J, Chen H. Navel orange peel essential oil inhibits the growth and progression of triple negative breast cancer. BMC Complement Med Ther 2024; 24:233. [PMID: 38877505 PMCID: PMC11177363 DOI: 10.1186/s12906-024-04525-y] [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: 10/30/2023] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Triple Negative Breast Cancer (TNBC) is a particular type of breast cancer with the highest mortality rate. Essential oils are concerned more and more as potential anti-cancer drugs. METHODS TNBC cells were treated with different concentrations of navel orange peel essential oil (NOPEO), and then a variety of experiments were performed to investigate the changes in the growth and progression of TNBC cells. MTT assay was performed to detect the proliferation of TNBC cells. The changes of cell cycle and apoptosis were analyzed by FACS. In order to explored the migration of TNBC cells, scratch wound assay was carried out. Western blotting and qPCR were used to examine the expression of proteins and mRNA of related genes. Furthermore, RNA-seq was used to analyze the altered genes and explored the possible signal pathway. RESULTS NOPEO demonstrated dose- and time-dependent suppression of TNBC cell growth. TNBC cells showed an increased percentage of G2/M-phase cells and the protein levels of CyclinB1 and CyclinD1 were decreased after NOPEO treatment. The apoptotic cells were increased in the NOPEO treated TNBC cells. The migration mobility was significantly inhibited by NOPEO. In total, 1376 genes were found to be up-regulated and 1335 genes were down-regulated after NOPEO treatment. According to KEGG and GO pathways, the differentially expressed genes were related to MAPK, Jak/stat and FoxQ signaling pathways. CONCLUSION This investigation explored the bio-activity and molecular mechanisms of NOPEO against TNBC cells. These results indicated that NOPEO could suppress TNBC growth and migration perhaps via the MAPK and Jak/stat signaling pathways, which may provide theoretical reference for anticancer drug development. NOPEO may be a potential natural product for the chemotherapeutic of TNBC.
Collapse
Affiliation(s)
- Chao Yang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China
| | - Wenwen Zhang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shi Xiang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Lai Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jiong Chun
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| | - Hui Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| |
Collapse
|
11
|
Wang YH, Gao P, Wang YQ, Xu LZ, Zeng KW, Tu PF. Small-molecule targeting PKM2 provides a molecular basis of lactylation-dependent fibroblast-like synoviocytes proliferation inhibition against rheumatoid arthritis. Eur J Pharmacol 2024; 972:176551. [PMID: 38570082 DOI: 10.1016/j.ejphar.2024.176551] [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: 10/19/2023] [Revised: 03/07/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Fibroblast-like synoviocytes (FLS) play an important role in rheumatoid arthritis (RA)-related swelling and bone damage. Therefore, novel targets for RA therapy in FLS are urgently discovered for improving pathologic phenomenon, especially joint damage and dyskinesia. Here, we suggested that pyruvate kinase M2 (PKM2) in FLS represented a pharmacological target for RA treatment by antimalarial drug artemisinin (ART). We demonstrated that ART selectively inhibited human RA-FLS and rat collagen-induced arthritis (CIA)-FLS proliferation and migration without observed toxic effects. In particular, the identification of targets revealed that PKM2 played a crucial role as a primary regulator of the cell cycle, leading to the heightened proliferation of RA-FLS. ART exhibited a direct interaction with PKM2, resulting in an allosteric modulation that enhances the lactylation modification of PKM2. This interaction further promoted the binding of p300, ultimately preventing the nuclear translocation of PKM2 and inducing cell cycle arrest at the S phase. In vivo, ART obviously suppressed RA-mediated synovial hyperplasia, bone damage and inflammatory response to further improve motor behavior in CIA-rats. Taken together, these findings indicate that directing interventions towards PKM2 in FLS could offer a hopeful avenue for pharmaceutical treatments of RA through the regulation of cell cycle via PKM2 lactylation.
Collapse
Affiliation(s)
- Yan-Hang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Peng Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Qi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lu-Zheng Xu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
12
|
Haake SM, Rios BL, Pozzi A, Zent R. Integrating integrins with the hallmarks of cancer. Matrix Biol 2024; 130:20-35. [PMID: 38677444 DOI: 10.1016/j.matbio.2024.04.003] [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: 01/09/2024] [Revised: 04/02/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
Epithelial cells adhere to a specialized extracellular matrix called the basement membrane which allows them to polarize and form epithelial tissues. The extracellular matrix provides essential physical scaffolding and biochemical and biophysical cues required for tissue morphogenesis, differentiation, function, and homeostasis. Epithelial cell adhesion to the extracellular matrix (i.e., basement membrane) plays a critical role in organizing epithelial tissues, separating the epithelial cells from the stroma. Epithelial cell detachment from the basement membrane classically results in death, though detachment or invasion through the basement membrane represents a critical step in carcinogenesis. Epithelial cells bind to the extracellular matrix via specialized matrix receptors, including integrins. Integrins are transmembrane receptors that form a mechanical linkage between the extracellular matrix and the intracellular cytoskeleton and are required for anchorage-dependent cellular functions such as proliferation, migration, and invasion. The role of integrins in the development, growth, and dissemination of multiple types of carcinomas has been investigated by numerous methodologies, which has led to great complexity. To organize this vast array of information, we have utilized the "Hallmarks of Cancer" from Hanahan and Weinberg as a convenient framework to discuss the role of integrins in the pathogenesis of cancers. This review explores this biology and how its complexity has impacted the development of integrin-targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Scott M Haake
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA.
| | - Brenda L Rios
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA
| | - Ambra Pozzi
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Roy Zent
- Department of Veterans Affairs, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA; Cancer Biology Program, Vanderbilt University, Nashville, TN, USA; Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
13
|
Mascanzoni F, Ayala I, Iannitti R, Luini A, Colanzi A. The Golgi checkpoint: Golgi unlinking during G2 is necessary for spindle formation and cytokinesis. Life Sci Alliance 2024; 7:e202302469. [PMID: 38479814 PMCID: PMC10941482 DOI: 10.26508/lsa.202302469] [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: 11/03/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
Entry into mitosis requires not only correct DNA replication but also extensive cell reorganization, including the separation of the Golgi ribbon into isolated stacks. To understand the significance of pre-mitotic Golgi reorganization, we devised a strategy to first block Golgi segregation, with the consequent G2-arrest, and then force entry into mitosis. We found that the cells forced to enter mitosis with an intact Golgi ribbon showed remarkable cell division defects, including spindle multipolarity and binucleation. The spindle defects were caused by reduced levels at the centrosome of the kinase Aurora-A, a pivotal spindle formation regulator controlled by Golgi segregation. Overexpression of Aurora-A rescued spindle formation, indicating a crucial role of the Golgi-dependent recruitment of Aurora-A at the centrosome. Thus, our results reveal that alterations of the pre-mitotic Golgi segregation in G2 have profound consequences on the fidelity of later mitotic processes and represent potential risk factors for cell transformation and cancer development.
Collapse
Affiliation(s)
- Fabiola Mascanzoni
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Roberta Iannitti
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
14
|
Ren J, Yao X, Yang M, Cheng S, Wu D, Xu K, Li R, Zhang H, Zhang D. Kinesin Family Member-18A (KIF18A) Promotes Cell Proliferation and Metastasis in Hepatocellular Carcinoma. Dig Dis Sci 2024; 69:1274-1286. [PMID: 38446308 PMCID: PMC11026273 DOI: 10.1007/s10620-024-08321-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/26/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND & AIMS Kinesin family member 18A (KIF18A) is notable for its aberrant expression across various cancer types and its pivotal role is driving cancer progression. In this study, we aim to investigate the intricate molecular mechanisms underlying the impact of KIF18A on the progression of HCC. METHODS Western blotting assays, a quantitative real-time PCR and immunohistochemical analyses were performed to quantitatively assess KIF18A expression in HCC tissues. We then performed genetic manipulations within HCC cells by silencing endogenous KIF18A using short hairpin RNA (shRNA) and introducing exogenous plasmids to overexpress KIF18A. We monitored cell progression, analyzed cell cycle and cell apoptosis and assessed cell migration and invasion both in vitro and in vivo. Moreover, we conducted RNA-sequencing to explore KIF18A-related signaling pathways utilizing Reactome and KEGG enrichment methods and validated these critical mediators in these pathways. RESULTS Analysis of the TCGA-LIHC database revealed pronounced overexpression of KIF18A in HCC tissues, the finding was subsequently confirmed through the analysis of clinical samples obtained from HCC patients. Notably, silencing KIF18A in cells led to an obvious inhibition of cell proliferation, migration and invasion in vitro. Furthermore, in subcutaneous and orthotopic xenograft models, suppression of KIF18A sgnificantly redudce tumor weight and the number of lung metastatic nodules. Mechanistically, KIF18A appears to facilitate cell proliferation by upregulating MAD2 and CDK1/CyclinB1 expression levels, with the activation of SMAD2/3 signaling contributing to KIF18A-driven metastasis. CONCLUSION Our study elucidates the molecular mechanism by which KIF18A mediates proliferation and metastasis in HCC cells, offering new insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Jihua Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Xinyan Yao
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Minli Yang
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Shengtao Cheng
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Daiqing Wu
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Kexin Xu
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Ranran Li
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Han Zhang
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Dapeng Zhang
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China.
- , Room 706, Chongyi Building, 1 Yixue Yuan Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
15
|
Herzog S, Fläschner G, Incaviglia I, Arias JC, Ponti A, Strohmeyer N, Nava MM, Müller DJ. Monitoring the mass, eigenfrequency, and quality factor of mammalian cells. Nat Commun 2024; 15:1751. [PMID: 38409119 PMCID: PMC10897412 DOI: 10.1038/s41467-024-46056-7] [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: 01/17/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024] Open
Abstract
The regulation of mass is essential for the development and homeostasis of cells and multicellular organisms. However, cell mass is also tightly linked to cell mechanical properties, which depend on the time scales at which they are measured and change drastically at the cellular eigenfrequency. So far, it has not been possible to determine cell mass and eigenfrequency together. Here, we introduce microcantilevers oscillating in the Ångström range to monitor both fundamental physical properties of the cell. If the oscillation frequency is far below the cellular eigenfrequency, all cell compartments follow the cantilever motion, and the cell mass measurements are accurate. Yet, if the oscillating frequency approaches or lies above the cellular eigenfrequency, the mechanical response of the cell changes, and not all cellular components can follow the cantilever motions in phase. This energy loss caused by mechanical damping within the cell is described by the quality factor. We use these observations to examine living cells across externally applied mechanical frequency ranges and to measure their total mass, eigenfrequency, and quality factor. The three parameters open the door to better understand the mechanobiology of the cell and stimulate biotechnological and medical innovations.
Collapse
Affiliation(s)
- Sophie Herzog
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Gotthold Fläschner
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland.
- Nanosurf AG, Gräubernstrasse 12, 4410, Liestal, Switzerland.
| | - Ilaria Incaviglia
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Javier Casares Arias
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Aaron Ponti
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Nico Strohmeyer
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Michele M Nava
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Klingelbergstrasse 48, 4056, Basel, Switzerland.
| |
Collapse
|
16
|
Bock F, Dong X, Li S, Viquez OM, Sha E, Tantengco M, Hennen EM, Plosa E, Ramezani A, Brown KL, Whang YM, Terker AS, Arroyo JP, Harrison DG, Fogo A, Brakebusch CH, Pozzi A, Zent R. Rac1 promotes kidney collecting duct repair by mechanically coupling cell morphology to mitotic entry. SCIENCE ADVANCES 2024; 10:eadi7840. [PMID: 38324689 PMCID: PMC10849615 DOI: 10.1126/sciadv.adi7840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 01/03/2024] [Indexed: 02/09/2024]
Abstract
Prolonged obstruction of the ureter, which leads to injury of the kidney collecting ducts, results in permanent structural damage, while early reversal allows for repair. Cell structure is defined by the actin cytoskeleton, which is dynamically organized by small Rho guanosine triphosphatases (GTPases). In this study, we identified the Rho GTPase, Rac1, as a driver of postobstructive kidney collecting duct repair. After the relief of ureteric obstruction, Rac1 promoted actin cytoskeletal reconstitution, which was required to maintain normal mitotic morphology allowing for successful cell division. Mechanistically, Rac1 restricted excessive actomyosin activity that stabilized the negative mitotic entry kinase Wee1. This mechanism ensured mechanical G2-M checkpoint stability and prevented premature mitotic entry. The repair defects following injury could be rescued by direct myosin inhibition. Thus, Rac1-dependent control of the actin cytoskeleton integrates with the cell cycle to mediate kidney tubular repair by preventing dysmorphic cells from entering cell division.
Collapse
Affiliation(s)
- Fabian Bock
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xinyu Dong
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shensen Li
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Olga M. Viquez
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric Sha
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew Tantengco
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth M. Hennen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Erin Plosa
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alireza Ramezani
- Interdisciplinary Center for Quantitative Modeling in Biology, University of California, Riverside, CA, USA
- Department of Physics and Astronomy, University of California, Riverside, CA, USA
| | - Kyle L. Brown
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Young Mi Whang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew S. Terker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan Pablo Arroyo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David G. Harrison
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Agnes Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cord H. Brakebusch
- Biotech Research Center, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Physiology and Molecular Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, TN, USA
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
17
|
Schmidt CJ, Stehbens SJ. Microtubule control of migration: Coordination in confinement. Curr Opin Cell Biol 2024; 86:102289. [PMID: 38041936 DOI: 10.1016/j.ceb.2023.102289] [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: 07/26/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/04/2023]
Abstract
The microtubule cytoskeleton has a well-established, instrumental role in coordinating cell migration. Decades of research has focused on understanding how microtubules couple intracellular trafficking with cortical targeting and spatial organization of signaling to facilitate locomotion. Movement in physically challenging environments requires coordination of forces generated by the actin cytoskeleton to drive cell shape changes, with microtubules acting to spatially regulate contractility. Recent work has demonstrated that the mechanical properties of microtubules are adaptive to stress, leading to a new understanding of their roles in cell migration. Herein we review new developments in how microtubules sense and adapt to changes in the physical properties of their environment during migration. We frame our discussion around our current understanding of how microtubules target cell-matrix adhesions, and their role in the spatiotemporal coordination of signaling to form mechano feedback loops. We expand on how these mechanisms may influence cell morphology in confined three-dimensional settings, and the importance of locally tuning the mechanical stability of polymers in response to mechanical cues. Finally, we discuss new roles for Golgi-derived microtubules in mechanosensing, and how preferential motor use may influence polymer stability to resist the physical constraints cells experience in confined environments.
Collapse
Affiliation(s)
- Christanny J Schmidt
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Samantha J Stehbens
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
18
|
Jones MJ, Jones MC. Cell cycle control by cell-matrix interactions. Curr Opin Cell Biol 2024; 86:102288. [PMID: 38056140 DOI: 10.1016/j.ceb.2023.102288] [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: 09/30/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
Cell adhesion to the extracellular matrix (ECM) is required for normal cell cycle progression and accurate cell division. However, how cell adhesion to the wide range of ECM proteins found in human tissues influences the cell cycle is not fully understood. The composition and physical properties of the ECM can have profound effects on cell proliferation but can also promote cell cycle exit and quiescence. Furthermore, during tumor development and progression, changes in the ECM can drive both cancer cell proliferation and dormancy. Cell-matrix adhesion is primarily sensed via integrin-associated adhesion complexes, which in turn are regulated by the cell cycle machinery. In particular, cyclin-dependent kinase 1 (CDK1) has been shown to play a crucial role in regulating adhesion complexes during interphase and entry into mitosis. These reciprocal links between cell cycle progression and cell-matrix interactions are now being identified.
Collapse
Affiliation(s)
- Michael J Jones
- Peninsula Medical School, Faculty of Health, Medicine, Dentistry and Human Sciences, University of Plymouth, PL6 8BU, United Kingdom
| | - Matthew C Jones
- Peninsula Medical School, Faculty of Health, Medicine, Dentistry and Human Sciences, University of Plymouth, PL6 8BU, United Kingdom.
| |
Collapse
|
19
|
Karbanová J, Deniz IA, Wilsch-Bräuninger M, de Sousa Couto RA, Fargeas CA, Santos MF, Lorico A, Corbeil D. Extracellular lipidosomes containing lipid droplets and mitochondria are released during melanoma cell division. Cell Commun Signal 2024; 22:57. [PMID: 38243233 PMCID: PMC10799373 DOI: 10.1186/s12964-024-01471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The incidence of melanoma is increasing worldwide. Since metastatic melanoma is highly aggressive, it is important to decipher all the biological aspects of melanoma cells. In this context, we have previously shown that metastatic FEMX-I melanoma cells release small (< 150 nm) extracellular vesicles (EVs) known as exosomes and ectosomes containing the stem (and cancer stem) cell antigenic marker CD133. EVs play an important role in intercellular communication, which could have a micro-environmental impact on surrounding tissues. RESULTS We report here a new type of large CD133+ EVs released by FEMX-I cells. Their sizes range from 2 to 6 µm and they contain lipid droplets and mitochondria. Real-time video microscopy revealed that these EVs originate from the lipid droplet-enriched cell extremities that did not completely retract during the cell division process. Once released, they can be taken up by other cells. Silencing CD133 significantly affected the cellular distribution of lipid droplets, with a re-localization around the nuclear compartment. As a result, the formation of large EVs containing lipid droplets was severely compromised. CONCLUSION Given the biochemical effect of lipid droplets and mitochondria and/or their complexes on cell metabolism, the release and uptake of these new large CD133+ EVs from dividing aggressive melanoma cells can influence both donor and recipient cells, and therefore impact melanoma growth and dissemination.
Collapse
Affiliation(s)
- Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, Dresden, 01307, Germany
| | - Ilker A Deniz
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, Dresden, 01307, Germany
| | - Michaela Wilsch-Bräuninger
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, Dresden, 01307, Germany
| | - Rita Alexandra de Sousa Couto
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany
- Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua de Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Christine A Fargeas
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, Dresden, 01307, Germany
| | - Mark F Santos
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV, 89014, USA
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV, 89014, USA.
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, Dresden, 01307, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, Dresden, 01307, Germany.
| |
Collapse
|
20
|
Shan Y, Zheng L, Zhang S, Qian B. Abnormal expression of FOXM1 in carcinogenesis of renal cell carcinoma: From experimental findings to clinical applications. Biochem Biophys Res Commun 2024; 692:149251. [PMID: 38056162 DOI: 10.1016/j.bbrc.2023.149251] [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: 09/12/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 12/08/2023]
Abstract
Renal cell carcinoma (RCC) is a prevalent malignancy within the genitourinary system. At present, patients with high-grade or advanced RCC continue to have a bleak prognosis. Mounting research have emphasized the significant involvement of Forkhead box M1 (FOXM1) in RCC development and progression. Therefore, it is imperative to consolidate the existing evidence regarding the contributions of FOXM1 to RCC tumorigenesis through a comprehensive review. This study elucidated the essential functions of FOXM1 in promoting RCC growth, invasion, and metastasis by regulating cell cycle progression, DNA repair, angiogenesis, and epithelial-mesenchymal transition (EMT). Also, FOXM1 might serve as a novel diagnostic and prognostic biomarker as well as a therapeutic target for RCC. Clinical findings demonstrated that the expression of FOXM1 was markedly upregulated in RCC samples, while a high level of FOXM1 was found to be associated with a poor overall survival rate of RCC. Furthermore, it is worth noting that FOXM1 may have a significant impact on the resistance of renal cell carcinoma (RCC) to radiotherapy. This observation suggests that inhibiting FOXM1 could be a promising strategy to impede the progression of RCC and enhance its sensitivity to radiotherapy. The present review highlighted the pivotal role of FOXM1 in RCC development. FOXM1 has the capacity to emerge as not only a valuable diagnostic and prognostic tool but also a viable therapeutic option for unresectable RCC.
Collapse
Affiliation(s)
- Yanmei Shan
- Department of Nephrology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, China
| | - Liying Zheng
- Postgraduate Department, First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Shilong Zhang
- Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Biao Qian
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China; Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
21
|
He T, Li C, Chen Q, Li R, Luo J, Mao J, Yang Z. Combined analysis of lncRNA and mRNA emphasizes the potential role of tryptophan-mediated regulation of muscle development in weaned piglets by lncRNA. J Anim Sci 2024; 102:skae264. [PMID: 39276131 PMCID: PMC11465388 DOI: 10.1093/jas/skae264] [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: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024] Open
Abstract
Pork is an important high-value protein source that fulfills the nutritional requirements for normal growth development, repair, and metabolism. Tryptophan (Trp), a crucial amino acid for piglet growth performance and muscle development, has an essential yet unclear regulatory mechanism. To investigate the biological basis of Trp regulation of piglet muscle development and identify the related regulatory pathways, we studied 20 weaned piglets. The piglets were divided into control (CON, 0.14% Trp) and high Trp (HT, 0.35% Trp) groups. They were fed with different Trp concentrations for 28 d, after which we collected the longissimus dorsi (LD) muscle for histomorphometric analysis and RNA extraction. Our results showed that the HT diet significantly increased the average daily weight gain, myocyte number, and muscle fiber density in weaned piglets. We then analyzed the differentially expressed (DE) genes in the LD muscle through RNA sequencing (RNA-seq). We identified 253 lncRNAs and 1,055 mRNAs mainly involved in myoblast proliferation and myofiber formation, particularly through the FoxO and AMPK signaling pathways and metabolism. Further analysis of the DE lncRNA targeting relationship and construction of a protein-protein interaction network resulted in the discovery of a novel lncRNA, XLOC_021675, or FRPMD, and elucidated its role in regulating piglet muscle development. Finally, we confirmed the RNA-seq results by reverse transcription polymerase chain reaction (RT-PCR). This study provides valuable insights into the regulatory mechanism of lncRNA-mediated Trp regulation of muscle development in weaned piglets offering a theoretical basis for optimizing piglet dietary ratios and enhancing pork production.
Collapse
Affiliation(s)
- Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Chenlei Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Qingyun Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ruiqian Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Ju Luo
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Jiani Mao
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
22
|
Li K, Huang Z, Liu C, Xu Y, Chen W, Shi L, Li C, Zhou F, Zhou F. Transcriptomic analysis of human pulmonary microvascular endothelial cells treated with LPS. Cell Signal 2023; 111:110870. [PMID: 37633475 DOI: 10.1016/j.cellsig.2023.110870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Acute respiratory distress syndrome (ARDS) has a rapid onset and progression, which lead to the severity and complexity of the primary disease and significantly increase the fatality rate of patients. Transcriptomics provides some ideas for clarifying the mechanism of ARDS, exploring prevention and treatment targets, and searching for related specific markers. In this study, RNA-Seq technology was used to observe the gene expression of human pulmonary microvascular endothelial cells (PMVECs) induced by LPS, and to excavate the key genes and signaling pathways in ARDS process. A total of 2300 up-regulated genes were detected, and a corresponding 1696 down-regulated genes were screened. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and protein-protein interaction (PPI) were also used for functional annotation of key genes. TFDP1 was identified as a cell cycle-dependent differentially expressed gene, and its reduced expression was verified in LPS-treated PMVECs and lung tissues of CLP-induced mice. In addition, the inhibition of TFDP1 on inflammation and apoptosis, and the promotion of proliferation were confirmed. The decreased expression of E2F1, Rb, CDK1 and the activation of MAPK signaling pathway were substantiated in the in vivo and in vitro models of ARDS. Moreover, SREBF1 has been demonstrated to be involved in cell cycle arrest in PMVECs by inhibiting CDK1. Our study shows that transcriptomics combined with basic research can broaden the investigation of ARDS mechanisms and may provide a basis for future mechanistic innovations.
Collapse
Affiliation(s)
- Kaili Li
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| | - Zuotian Huang
- Department of Hepatobiliary Pancreatic Tumor Center, Chongqing University Cancer Hospital, 400030 Chongqing Municipality, China
| | - Chang Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| | - Yuanyuan Xu
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Wei Chen
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Lu Shi
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Can Li
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Fawei Zhou
- Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Fachun Zhou
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China; Department of Critical Care Medicine, the First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China.
| |
Collapse
|
23
|
Wang Y, Tong X, Shi X, Keswani T, Chatterjee E, Chen L, Li G, Lee K, Guo T, Yu Y. Chiral Cell Nanomechanics Originated in Clockwise/Counterclockwise Biofunctional Microarrays to Govern the Nuclear Mechanotransduction of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48038-48049. [PMID: 37812566 DOI: 10.1021/acsami.3c11188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Cell chirality is extremely important for the evolution of cell morphogenesis to manipulate cell performance due to left-right asymmetry. Although chiral micro- and nanoscale biomaterials have been developed to regulate cell functions, how cell chirality affects cell nanomechanics to command nuclear mechanotransduction was ambiguous. In this study, chiral engineered microcircle arrays were prepared by photosensitive cross-linking synthesis on cell culture plates to control the clockwise/counterclockwise geometric topology of stem cells. Asymmetric focal adhesion and cytoskeleton structures could induce chiral cell nanomechanics measured by atomic force microscopy (AFM) nanoindentation in left-/right-handed stem cells. Cell nanomechanics could be enhanced when the construction of mature focal adhesion and the assembly of actin and myosin cytoskeletons were well organized in chiral engineered stem cells. Curvature angles had a negative effect on cell nanomechanics, while cell chirality did not change cytoskeletal mechanics. The biased cytoskeleton tension would engender different nuclear mechanotransductions by yes-associated protein (YAP) evaluation. The chiral stimuli were delivered into the nuclei to oversee nuclear behaviors. A strong cell modulus could activate high nuclear DNA synthesis activity by mechanotransduction. The results will bring the possibility of understanding the interplay of chiral cell nanomechanics and mechanotransduction in nanomedicines and biomaterials.
Collapse
Affiliation(s)
- Yongtao Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Xiaolan Tong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Xiaohui Shi
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, 333 Nan Chen Road, Shanghai 200444, China
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lei Chen
- Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Guoping Li
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, United States
| | - Kyubae Lee
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tao Guo
- Department of Orthopaedics, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Yan Yu
- Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Spine Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
24
|
Zeng K, Li W, Wang Y, Zhang Z, Zhang L, Zhang W, Xing Y, Zhou C. Inhibition of CDK1 Overcomes Oxaliplatin Resistance by Regulating ACSL4-mediated Ferroptosis in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301088. [PMID: 37428466 PMCID: PMC10477855 DOI: 10.1002/advs.202301088] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/31/2023] [Indexed: 07/11/2023]
Abstract
Oxaliplatin is a widely used chemotherapy drug for patients with advanced colorectal cancer (CRC); however, frequent drug resistance limits its therapeutic efficacy in patients. Here, this work identifies cyclin-dependent kinase 1 (CDK1) as a critical contributor to oxaliplatin resistance via in vitro and in vivo CRISPR/Cas9 screening. CDK1 is highly expressed in oxaliplatin-resistant cells and tissues due to the loss of N6-methyladenosine modification. Genetic and pharmacological blockade of CDK1 restore the susceptibility of CRC cells to oxaliplatin in vitro and in cell/patient-derived xenograft models. Mechanistically, CDK1 directly binds to and phosphorylates Acyl-CoA synthetase long-chain family 4 (ACSL4) at S447, followed by recruitment of E3 ubiquitin ligase UBR5 and polyubiquitination of ACSL4 at K388, K498, and K690, which leads to ACSL4 protein degradation. Reduced ACSL4 subsequently blocks the biosynthesis of polyunsaturated fatty acid containing lipids, thereby inhibiting lipid peroxidation and ferroptosis, a unique iron-dependent form of oxidative cell death. Moreover, treatment with a ferroptosis inhibitor nullifies the enhancement of CRC cell sensitivity to oxaliplatin by CDK1 blockade in vitro and in vivo. Collectively, the findings indicate that CDK1 confers oxaliplatin resistance to cells by suppressing ferroptosis. Therefore, administration of a CDK1 inhibitor may be an attractive strategy to treat patients with oxaliplatin-resistant CRC.
Collapse
Affiliation(s)
- Kaixuan Zeng
- Precision Medical Research Institutethe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710000China
| | - Weihao Li
- Department of Colorectal SurgerySun Yat‐sen University Cancer CenterGuangzhou510060China
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yue Wang
- Department of Gastroenterologythe First Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Zifei Zhang
- IIT Project Management Officethe First Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Linjie Zhang
- Department of Colorectal SurgerySun Yat‐sen University Cancer CenterGuangzhou510060China
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Weili Zhang
- Department of Colorectal SurgerySun Yat‐sen University Cancer CenterGuangzhou510060China
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| | - Yue Xing
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationMedical Research CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
- Breast Tumor CenterSun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhou510120China
| | - Chi Zhou
- Department of Colorectal SurgerySun Yat‐sen University Cancer CenterGuangzhou510060China
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
25
|
Ganguli S, Wyatt T, Nyga A, Lawson RH, Meyer T, Baum B, Matthews HK. Oncogenic Ras deregulates cell-substrate interactions during mitotic rounding and respreading to alter cell division orientation. Curr Biol 2023; 33:2728-2741.e3. [PMID: 37343559 PMCID: PMC7614879 DOI: 10.1016/j.cub.2023.05.061] [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: 01/06/2023] [Revised: 04/21/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023]
Abstract
Oncogenic Ras has been shown to change the way cancer cells divide by increasing the forces generated during mitotic rounding. In this way, RasV12 enables cancer cells to divide across a wider range of mechanical environments than normal cells. Here, we identify a further role for oncogenic Ras-ERK signaling in division by showing that RasV12 expression alters the shape, division orientation, and respreading dynamics of cells as they exit mitosis. Many of these effects appear to result from the impact of RasV12 signaling on actomyosin contractility, because RasV12 induces the severing of retraction fibers that normally guide spindle positioning and provide a memory of the interphase cell shape. In support of this idea, the RasV12 phenotype is reversed by inhibition of actomyosin contractility and can be mimicked by the loss of cell-substrate adhesion during mitosis. Finally, we show that RasV12 activation also perturbs division orientation in cells cultured in 2D epithelial monolayers and 3D spheroids. Thus, the induction of oncogenic Ras-ERK signaling leads to rapid changes in division orientation that, along with the effects of RasV12 on cell growth and cell-cycle progression, are likely to disrupt epithelial tissue organization and contribute to cancer dissemination.
Collapse
Affiliation(s)
- Sushila Ganguli
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Tom Wyatt
- Laboratoirè Matiere et Systèmes Complexes, Université Paris Diderot, 10 rue Alice Domon et Léonie Duquet, Bâtiment Condorcet, 75013 Paris, France
| | - Agata Nyga
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rachel H Lawson
- School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Tim Meyer
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Buzz Baum
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Helen K Matthews
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; School of Biosciences, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| |
Collapse
|
26
|
Bachmann M, Su B, Rahikainen R, Hytönen VP, Wu J, Wehrle-Haller B. ConFERMing the role of talin in integrin activation and mechanosignaling. J Cell Sci 2023; 136:jcs260576. [PMID: 37078342 PMCID: PMC10198623 DOI: 10.1242/jcs.260576] [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: 04/21/2023] Open
Abstract
Talin (herein referring to the talin-1 form), is a cytoskeletal adapter protein that binds integrin receptors and F-actin, and is a key factor in the formation and regulation of integrin-dependent cell-matrix adhesions. Talin forms the mechanical link between the cytoplasmic domain of integrins and the actin cytoskeleton. Through this linkage, talin is at the origin of mechanosignaling occurring at the plasma membrane-cytoskeleton interface. Despite its central position, talin is not able to fulfill its tasks alone, but requires help from kindlin and paxillin to detect and transform the mechanical tension along the integrin-talin-F-actin axis into intracellular signaling. The talin head forms a classical FERM domain, which is required to bind and regulate the conformation of the integrin receptor, as well as to induce intracellular force sensing. The FERM domain allows the strategic positioning of protein-protein and protein-lipid interfaces, including the membrane-binding and integrin affinity-regulating F1 loop, as well as the interaction with lipid-anchored Rap1 (Rap1a and Rap1b in mammals) GTPase. Here, we summarize the structural and regulatory features of talin and explain how it regulates cell adhesion and force transmission, as well as intracellular signaling at integrin-containing cell-matrix attachment sites.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| | - Baihao Su
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA
| | - Rolle Rahikainen
- Faculty of Medicine and Health Technology, Arvo Ylpön katu 34, Tampere University, FI-33520 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Arvo Ylpön katu 34, Tampere University, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Molecular Therapeutics Program, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111, USA
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, Centre Médical Universitaire, 1211 Geneva 4, Switzerland
| |
Collapse
|
27
|
Huber M, Casares-Arias J, Fässler R, Müller DJ, Strohmeyer N. In mitosis integrins reduce adhesion to extracellular matrix and strengthen adhesion to adjacent cells. Nat Commun 2023; 14:2143. [PMID: 37059721 PMCID: PMC10104879 DOI: 10.1038/s41467-023-37760-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
To enter mitosis, most adherent animal cells reduce adhesion, which is followed by cell rounding. How mitotic cells regulate adhesion to neighboring cells and extracellular matrix (ECM) proteins is poorly understood. Here we report that, similar to interphase, mitotic cells can employ integrins to initiate adhesion to the ECM in a kindlin- and talin-dependent manner. However, unlike interphase cells, we find that mitotic cells cannot engage newly bound integrins to actomyosin via talin or vinculin to reinforce adhesion. We show that the missing actin connection of newly bound integrins leads to transient ECM-binding and prevents cell spreading during mitosis. Furthermore, β1 integrins strengthen the adhesion of mitotic cells to adjacent cells, which is supported by vinculin, kindlin, and talin1. We conclude that this dual role of integrins in mitosis weakens the cell-ECM adhesion and strengthens the cell-cell adhesion to prevent delamination of the rounding and dividing cell.
Collapse
Affiliation(s)
- Maximilian Huber
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland
| | - Javier Casares-Arias
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Daniel J Müller
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland.
| | - Nico Strohmeyer
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, 4058, Basel, Switzerland.
| |
Collapse
|
28
|
Phosphorylation of VP1 Mediated by CDK1-Cyclin B1 Facilitates Infectious Bursal Disease Virus Replication. J Virol 2023; 97:e0194122. [PMID: 36602364 PMCID: PMC9888224 DOI: 10.1128/jvi.01941-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Infectious bursal disease virus (IBDV) is a double-stranded RNA (dsRNA) virus belonging to the genus Avibirnavirus in the family Birnaviridae. It can cause serious failure of vaccination in young poultry birds with impaired immune systems. Post-translational modifications of the VP1 protein are essential for viral RNA transcription, genome replication, and viral multiplication. Little information is available so far regarding the exact mechanism of phosphorylation of IBDV VP1 and its significance in the viral life cycle. Here, we provide several lines of evidence that the cyclin-dependent kinase 1 (CDK1)-cyclin B1 complex phosphorylates VP1, which facilitates viral replication. We show that the CDK1-cyclin B1 specifically interacts with VP1 and phosphorylates VP1 on the serine 7 residue, located in the N-terminal 7SPAQ10 region, which follows the optimal phosphorylation motif of CDK1, p-S/T-P. Additionally, IBDV infection drives the cytoplasmic accumulation of CDK1-cyclin B1, which co-localizes with VP1, supporting the kinase activity of CDK1-cyclin B1. Treatment with CDK1 inhibitor RO3306 and knockdown of CDK1-cyclin B1 severely disrupts the polymerase activity of VP1, resulting in diminished viral replication. Moreover, the replication of S7A mutant recombinant IBDV was significantly decreased compared to that of wild-type (WT) IBDV. Thus, CDK1-cyclin B1 is a crucial enzyme which phosphorylates IBDV VP1 on serine 7, which is necessary both for the polymerase activity of VP1 and for viral replication. IMPORTANCE Infectious bursal disease virus still poses a great economic threat to the global poultry farming industry. Detailed information on the steps of viral genome replication is essential for the development of antiviral therapeutics. Phosphorylation is a common post-translational modification in several viral proteins. There is a lack of information regarding the significance of VP1 phosphorylation and its role in modulating the viral life cycle. In this study, we found that CDK1-cyclin B1 accumulates in the cytoplasm and phosphorylates VP1 on serine 7. The presence of a CDK1 inhibitor and the silencing of CDK1-cyclin B1 decrease IBDV replication. The mutation of VP1 serine 7 to alanine reduces VP1 polymerase activity, disrupting the viral life cycle, which suggests that this residue serves an essential function. Our study offers novel insights into the regulatory mechanism of VP1 phosphorylation.
Collapse
|
29
|
Cell Cycle Regulation by Integrin-Mediated Adhesion. Cells 2022; 11:cells11162521. [PMID: 36010598 PMCID: PMC9406542 DOI: 10.3390/cells11162521] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Cell cycle and cell adhesion are two interdependent cellular processes regulating each other, reciprocally, in every cell cycle phase. The cell adhesion to the extracellular matrix (ECM) via integrin receptors triggers signaling pathways required for the cell cycle progression; the passage from the G1 to S phase and the completion of cytokinesis are the best-understood events. Growing evidence, however, suggests more adhesion-dependent regulatory aspects of the cell cycle, particularly during G2 to M transition and early mitosis. Conversely, the cell cycle machinery regulates cell adhesion in manners recently shown driven mainly by cyclin-dependent kinase 1 (CDK1). This review summarizes the recent findings regarding the role of integrin-mediated cell adhesion and its downstream signaling components in regulating the cell cycle, emphasizing the cell cycle progression through the G2 and early M phases. Further investigations are required to raise our knowledge about the molecular mechanisms of crosstalk between cell adhesion and the cell cycle in detail.
Collapse
|
30
|
Zuidema A, Wang W, Kreft M, Bleijerveld OB, Hoekman L, Aretz J, Böttcher RT, Fässler R, Sonnenberg A. Molecular determinants of αVβ5 localization in flat clathrin lattices: Role of αVβ5 in cell adhesion and proliferation. J Cell Sci 2022; 135:275569. [PMID: 35532004 PMCID: PMC9234671 DOI: 10.1242/jcs.259465] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 04/20/2022] [Indexed: 11/20/2022] Open
Abstract
The vitronectin receptor integrin αVβ5 can reside in two distinct adhesion structures: focal adhesions (FAs) and flat clathrin lattices (FCLs). Here we investigated the mechanism that regulates the subcellular distribution of β5 in keratinocytes and show that β5 has approximately 7- and 5-fold higher affinity for the clathrin adaptors ARH and Numb, respectively, than for talin; all proteins that bind to the membrane-proximal NPxY motif of the β5 cytoplasmic domain. Using mass spectrometry, we identified β5 interactors including the Rho GEFs p115Rho-GEF and GEF-H1, and the serine protein kinase MARK2; depletion of which diminishes the clustering of β5 in FCLs. Substitution of two serines (S759/762) in the β5 cytoplasmic domain with phospho-mimetic glutamates causes a shift in the localization of β5 from FAs into FCLs without affecting the interactions with MARK2, p115Rho-GEF or GEF-H1. Instead, we demonstrate that changes in the actomyosin-based cellular contractility by ectopic expression of activated Rho or disruption of microtubules regulates β5 localization. Finally, we present evidence that β5 in either FAs or FCLs functions to promote adhesion to vitronectin, cell spreading, and proliferation.
Collapse
Affiliation(s)
- Alba Zuidema
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Wei Wang
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | - Maaike Kreft
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| | | | - Liesbeth Hoekman
- Proteomics Facility, The Netherlands Cancer Institute, The Netherlands
| | - Jonas Aretz
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, The Netherlands
| | - Ralph T. Böttcher
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, The Netherlands
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, The Netherlands
| | - Arnoud Sonnenberg
- Division of Cell Biology I, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam 1066 CX, The Netherlands
| |
Collapse
|