1
|
Wu T, Li W, Zhuang L, Liu J, Wang P, Gu Y, Liu Y, Yu Y. Deficiency of Aging-Related Gene Chitinase-Like 4 Impairs Olfactory Epithelium Homeostasis. Cell Prolif 2025:e70055. [PMID: 40389328 DOI: 10.1111/cpr.70055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/02/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
Mammalian olfactory epithelium (OE) undergoes consistent self-renewal throughout life. In OE homeostasis, globose basal cells (GBCs) contribute to the generation of olfactory sensory neurons (OSNs) to replace old ones. Chitinase-like 4 (Chil4), a chitinase-like protein expressed in supporting cells, plays a critical role in OE regeneration, while its role in tissue homeostasis is still elusive. Here, we found that Chil4 is upregulated in the aged OE. Deletion of Chil4 leads to a reduction in the number of GBCs and immature OSNs (iOSNs). Chil4-/- GBCs show attenuation in cell cycle progression and an aberrant expression pattern of cell-cycle-related genes such as Cdk1. Chil4 deletion causes loss of a specific subcluster of GAP43+ iOSNs expressing Cebpb, Nqo1 and low level of mature OSN (mOSN) marker Stoml3 (iOSN_CeStLNq), potentially suggesting a transitional state between immature and mature neurons. Chil4 knockout induces inflammatory activation in Iba1+ microglia (MG)-like cells in the OE. Chil4 downregulation in aged organoids reduced the number of mature sensory neurons, suggesting a necessary role of Chil4 in maintaining neuronal generation in the aged OE. Collectively, these observations reveal a previously unidentified function of Chil4, establishing the cellular mechanism underlying OE homeostasis.
Collapse
Affiliation(s)
- Tingting Wu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Weihao Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Olfactory Disorder Diagnosis and Treatment Center, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Liujing Zhuang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Jinxia Liu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Ye Gu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Yongliang Liu
- Department of Otolaryngology, Zibo Central Hospital, Zibo, Shandong, China
| | - Yiqun Yu
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Olfactory Disorder Diagnosis and Treatment Center, Eye & ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Shin YZ, Yum YA, Bae ES, Jarhad DB, Aswar VR, Tripathi SK, Kwon EJ, Kim YJ, Kim M, Lee SK, Jeong LS, Cha HJ. Targeting HASPIN in gemcitabine-resistant pancreatic cancer cells by lead optimization of thioadenosine analogue. Biomed Pharmacother 2025; 188:118135. [PMID: 40378773 DOI: 10.1016/j.biopha.2025.118135] [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: 03/17/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Despite multiple kinase inhibitors having been developed for cancer therapy, mitotic kinases remain difficult to target with small molecules due to severe adverse effects on proliferating normal cells. Recently, HASPIN, a mitotic kinase responsible for histone H3 phosphorylation, has emerged as a promising cancer-specific target. In this study, we synthesized a novel thioadenosine analogue, LJ5157, based on the structure of the previously developed HASPIN inhibitor LJ4827. In silico transcriptome analysis of pancreatic cancer patient data from The Cancer Genome Atlas identified HASPIN as not only a cancer-specific target but also a potential key player in overcoming gemcitabine resistance. To evaluate the therapeutic potential of LJ5157, we tested its efficacy in pancreatic cancer cells, particularly gemcitabine-resistant Panc-1 (GR) cells. The inhibitor exhibited potent anti-cancer activity, effectively suppressing the growth of GR cells, which showed more dysregulated cell cycle progression and greater proportion of polyploid cells compared to wild-type Panc-1 cells. Furthermore, it demonstrated superior efficacy in reducing the mitotic population of polyploid GR cells, which correlated with significant tumor growth inhibition in a GR-cell-derived xenograft model. Further optimization of LJ4827 led to development of LJ5242, an analogue with enhanced selectivity for HASPIN and improved cell cycle inhibitory potency. These findings highlight HASPIN inhibition as a promising strategy for targeting chemoresistant pancreatic cancer and further identify thioadenosine as a valuable pharmacophore for developing clinically viable HASPIN inhibitors.
Collapse
Affiliation(s)
- Yoon-Ze Shin
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yun A Yum
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Dnyandev B Jarhad
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Vikas R Aswar
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | | | - Eun-Ji Kwon
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yun-Jeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Minjae Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Natural Products Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Lak-Shin Jeong
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Future Medicine Co., Ltd, Seongnam, Gyeonggi-do, Republic of Korea.
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Hao Q, Zhao W, Li Z, Lai Y, Wang Y, Yang Q, Zhang L. Combination therapy and dual-target inhibitors based on cyclin-dependent kinases (CDKs): Emerging strategies for cancer therapy. Eur J Med Chem 2025; 289:117465. [PMID: 40037064 DOI: 10.1016/j.ejmech.2025.117465] [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: 01/04/2025] [Revised: 02/22/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Cyclin-dependent kinases (CDKs) are pivotal regulators of the cell cycle and transcriptional machinery, making them attractive targets for cancer therapy. While CDK inhibitors have demonstrated promising clinical outcomes, they also face challenges in enhancing efficacy, particularly in overcoming drug resistance. Combination therapies have emerged as a key strategy to augment the effectiveness of CDK inhibitors when used alongside other kinase inhibitors or non-kinase-targeted agents. Dual-target inhibitors that simultaneously inhibit CDKs and other oncogenic drivers are gaining attention, offering novel avenues to optimize cancer therapy. Based on the structural characterization and biological functions of CDKs, this review comprehensively examines the structure-activity relationship (SAR) of existing dual-target CDK inhibitors from a drug design perspective. We also thoroughly investigate the preclinical studies and clinical translational potential of combination therapies and dual-target inhibitors. Tailoring CDK inhibitors to specific cancer subtypes and therapeutic settings will inspire innovative approaches for the next generation of CDK-related therapies, ultimately improving patient survival.
Collapse
Affiliation(s)
- Qi Hao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Wenzhe Zhao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yue Lai
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Yan Wang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Qianqian Yang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China; Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
4
|
Deshpande A, Mahale S, Kanduri C. Beyond the Transcript: Translating Non-Coding RNAs and Their Impact on Cellular Regulation. Cancers (Basel) 2025; 17:1555. [PMID: 40361481 PMCID: PMC12071610 DOI: 10.3390/cancers17091555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Non-coding RNAs (ncRNAs) constitute the majority of the human transcriptome and play diverse structural, catalytic, and regulatory roles. The ability of ncRNAs to be translated into functional peptides and microproteins expands our understanding of their regulatory potential beyond their established non-coding functions. Our comprehensive search identified 86 translating "non-coding" RNAs. While translating ncRNAs have traditionally been categorized as "peptide-encoding", in this study, we introduce a novel classification based on amino acid length, distinguishing their products as ncRNA encoded peptides (ncRNA-PEPs), which are less than 60 amino acids, or ncRNA encoded microproteins (ncRNA-MPs) ranging from 61 to 200 amino acids. These peptides and microproteins act as co-regulators in cell signaling, transcriptional regulation, and protein complex assembly, playing a role in both health and disease. We outline the molecular pathways by which ncRNA-PEPs and ncRNA-MPs could govern cell cycle progression, highlighting their influence on cell cycle transitions, oncogenic and tumor suppressor pathways, metabolic homeostasis, autophagy, and on key cell cycle regulators like PCNA, Rad18, and CDK-cyclin complexes. Furthermore, we highlight recent advancements in their detection and characterization, exploring their evolutionary origins, species-specific conservation, and potential therapeutic applications. Our findings underscore the emerging significance of ncRNA-PEPs and ncRNA-MPs as integral regulators of cellular processes, highlighting their functional versatility and opening promising avenues for further research and potential therapeutic applications.
Collapse
Affiliation(s)
| | | | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden; (A.D.); (S.M.)
| |
Collapse
|
5
|
Le Hir-Reynaud E, Soubise B, Mendoza AM, Konan C, Commet S, Gueganic N, Tous C, Corcos L, Douet-Guilbert N, Troadec MB. RBM22-depletion delays progression through all steps of cell cycle and increases ploidy in myeloid cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119965. [PMID: 40268057 DOI: 10.1016/j.bbamcr.2025.119965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
RNA-Binding Motif 22 (RBM22) is a splicing factor and a transcription regulator that plays important roles in cancer. Our goal was to document further the implication of RBM22 in cell cycle progression. Using normal human haematopoietic stem and progenitor cells and myeloid cell lines (MDS-L, HL-60), we demonstrated that RBM22 depletion reduces proliferation by delaying the progression of the G1-phase, S-phase and G2/M phase. RBM22 depletion alters mitosis, generating endomitosis and alters megakaryocyte differentiation. Altogether, we propose, for the first time, RBM22 as an essential actor of the cell cycle regulation in human haematopoietic stem and progenitor cells and myeloid cells. We demonstrated that RBM22 alteration is partially responsible for the phenotype of cytopenia of myeloid cell lineages observed in myelodysplastic syndromes (MDS) with a partial deletion of chromosome 5 (MDS with del(5q)) where one allele of RBM22 is lost. We hypothesise that the impact of RBM22 on cell cycle progression could explain some phenotypic features of other cancers.
Collapse
Affiliation(s)
| | - Benoît Soubise
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France
| | | | - Cassandra Konan
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Séverine Commet
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, service de génétique, laboratoire de génétique chromosomique, Brest, France
| | - Nadia Gueganic
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, service de génétique, laboratoire de génétique chromosomique, Brest, France
| | - Corinne Tous
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, service de génétique, laboratoire de génétique chromosomique, Brest, France
| | - Laurent Corcos
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Nathalie Douet-Guilbert
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, service de génétique, laboratoire de génétique chromosomique, Brest, France
| | - Marie-Bérengère Troadec
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, service de génétique, laboratoire de génétique chromosomique, Brest, France.
| |
Collapse
|
6
|
Kwiatkowski N, Liang T, Sha Z, Collier PN, Yang A, Sathappa M, Paul A, Su L, Zheng X, Aversa R, Li K, Mehovic R, Kolodzy C, Breitkopf SB, Chen D, Howarth CL, Yuan K, Jo H, Growney JD, Weiss M, Williams J. CDK2 heterobifunctional degraders co-degrade CDK2 and cyclin E resulting in efficacy in CCNE1-amplified and overexpressed cancers. Cell Chem Biol 2025; 32:556-569.e24. [PMID: 40250405 DOI: 10.1016/j.chembiol.2025.03.006] [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: 09/11/2024] [Revised: 01/27/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025]
Abstract
CCNE1 amplification drives aberrant CDK2-cyclin E1 activity in cancer. Despite activity of CDK2 inhibitors, their therapeutic margins are limited by poor CDK selectivity. We developed a degrader with high selectivity for CDK2 over CDK1 that also unexpectedly led to cyclin E1 degradation and potent and complete suppression of RB phosphorylation at concentrations with low CDK2 occupancy and negligible CDK1 degradation. Co-depletion of CDK2 and cyclin E1 also resensitized palbociclib-adapted breast cancer cells to cell cycle blockade. Overall, the improved potency and selectivity of the degrader for CDK2 over small-molecule inhibitors drives antiproliferative activity with greater specificity for CCNE1amp cancer cells and RB dependency. Using an orally administered degrader, we demonstrate deep and sustained RB pathway suppression, which is needed to induce stasis in CCNE1amp tumors. These results highlight the potential of this modality to target CDK2 potently and selectivity in this biomarker-defined patient population with high unmet need.
Collapse
Affiliation(s)
- Nicholas Kwiatkowski
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Tong Liang
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Zhe Sha
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Philip N Collier
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Annan Yang
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Murugappan Sathappa
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Atanu Paul
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Lijing Su
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Xiaozhang Zheng
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Robert Aversa
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Kunhua Li
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Revonda Mehovic
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Christina Kolodzy
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Susanne B Breitkopf
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Dapeng Chen
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Charles L Howarth
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Karen Yuan
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Hakryul Jo
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Joseph D Growney
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Matthew Weiss
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Juliet Williams
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA.
| |
Collapse
|
7
|
Camlin NJ. Protein-targeting reverse genetic approaches: the future of oocyte and preimplantation embryo research. Mol Hum Reprod 2025; 31:gaaf008. [PMID: 40100642 PMCID: PMC12000532 DOI: 10.1093/molehr/gaaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Reverse genetic approaches are the standard in molecular biology to determine a protein's function. Traditionally, nucleic acid targeting via gene knockout (DNA) and knockdown (RNA) has been the method of choice to remove proteins-of-interest. However, the nature of mammalian oocyte maturation and preimplantation embryo development can make nucleic acid-targeting approaches difficult. Gene knockout allows time for compensatory mechanisms and secondary phenotypes to develop which can make interpretation of a protein's function difficult. Furthermore, genes can be essential for animal and/or oocyte survival, and therefore, gene knockout is not always a viable approach to investigate oocyte maturation and preimplantation embryo development. Conversely, RNA-targeting approaches, i.e. RNA interference (RNAi) and morpholinos, rely on protein half-life and therefore are unable to knockdown every protein-of-interest. An increasing number of reverse genetic approaches that directly target proteins have been developed to overcome the limitations of nucleic acid-based approaches, including Trim-Away and auxin-inducible degradation. These protein-targeting approaches give researchers exquisite and fast control of protein loss. This review will discuss how Trim-Away and auxin-inducible degradation can overcome many of the challenges of nucleic acid-based reverse genetic approaches. Furthermore, it highlights the unique research opportunities these approaches afford, such as targeting post-translationally modified proteins.
Collapse
Affiliation(s)
- Nicole J Camlin
- Cell and Molecular Biology, School of Biological, Environmental and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, USA
| |
Collapse
|
8
|
Wetherall B, Bulmer D, Sarginson A, Thomas C, Madgwick S. SGO2 does not play an essential role in separase inhibition during meiosis I in mouse oocytes. PLoS Biol 2025; 23:e3003131. [PMID: 40267054 PMCID: PMC12017502 DOI: 10.1371/journal.pbio.3003131] [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: 02/03/2025] [Accepted: 03/25/2025] [Indexed: 04/25/2025] Open
Abstract
During meiosis I in oocytes, anaphase is triggered by deactivation of cyclin B1-CDK1 and activation of separase. Active separase plays an essential role in cleaving cohesin rings that hold homologous chromosomes together. Critically, separase must be inhibited until all chromosomes are aligned and the cell is prepared for anaphase I. Inhibition can be mediated through the binding of separase to either securin or cyclin B1-CDK1. The relative contribution of each inhibitory pathway varies depending on cell type. Recently, shugoshin-2 (SGO2) has also been shown to inhibit separase in mitotic cells. Here, we used a separase biosensor and perturbed the three inhibitory pathways during meiosis I in mouse oocytes. We show that inhibition mediated by either securin or cyclin B1-CDK1, but not SGO2, is independently sufficient to suppress separase activity. However, when both the securin and cyclin B1-CDK1 inhibitory pathways are perturbed together, separase activity begins prematurely, resulting in gross segregation defects. Furthermore, we characterized SGO2 destruction dynamics and concluded that it is not an essential separase inhibitor in mouse oocytes. The existence of multiple separase inhibitory pathways highlights the critical importance of tightly regulated separase activity during this unique and challenging cell division.
Collapse
Affiliation(s)
- Benjamin Wetherall
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - David Bulmer
- Bioimaging Unit, Faculty of Medical Sciences, Newcastle University, Newcastle, United Kingdom
| | - Alexandra Sarginson
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| | - Christopher Thomas
- IBDM—Institut de Biologie du Développement de Marseille, CNRS—UMR 7288, Aix-Marseille Université, Marseille, France
| | - Suzanne Madgwick
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle, United Kingdom
| |
Collapse
|
9
|
Choi W, Park DJ, Dorschner RA, Nakatsutsumi K, Yi M, Eliceiri BP. CDK1-loaded extracellular vesicles promote cell cycle to reverse impaired wound healing in diabetic obese mice. Mol Ther 2025; 33:1118-1133. [PMID: 39865653 PMCID: PMC11897770 DOI: 10.1016/j.ymthe.2025.01.039] [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: 08/26/2024] [Revised: 12/30/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
Small extracellular vesicles (sEVs) mediate intercellular signaling to coordinate the proliferation of cell types that promote re-epithelialization of skin following injury. Cyclin-dependent kinase 1 (CDK1) drives cell division and is a key regulator of entry to the cell cycle. To understand the potential of sEV-mediated delivery of CDK1 to reverse impaired wound healing, we generated CDK1-loaded sEVs (CDK1-sEVs) and evaluated their ability to mediate cell proliferation, re-epithelialization, and downstream signaling responses in the wound bed. We found that treatment of human keratinocytes with CDK1-sEVs increased phosphorylation of the CDK1 target, eukaryotic translation inhibition factor 4E-binding protein 1 (4E-BP1), and histone H3 within 24 h via AKT and ERK phosphorylation, driving increased proliferation and cell migration. Treatment of the wound bed of diabetic obese mice, a model of delayed wound healing, with a single dose of CDK1-sEVs accelerated wound closure, increased re-epithelialization, and promoted the proliferation of keratinocytes. These studies show that delivery of CDK1 by sEVs can stimulate selective and transient proliferation of cell types that increase re-epithelialization and promote proliferation of keratinocytes to accelerate wound healing.
Collapse
Affiliation(s)
- Wooil Choi
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dong Jun Park
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert A Dorschner
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Keita Nakatsutsumi
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michelle Yi
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian P Eliceiri
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA; Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
10
|
Chen W, Zhuang X, Chen Y, Yang H, Shen L, Feng S, Min W, Yuan K, Yang P. Recent advances in regulating the cell cycle through inhibiting CDKs for cancer treatment. Chin J Nat Med 2025; 23:286-298. [PMID: 40122659 DOI: 10.1016/s1875-5364(25)60846-6] [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: 02/26/2024] [Revised: 06/11/2024] [Accepted: 06/29/2024] [Indexed: 03/25/2025]
Abstract
The inhibition of cyclin-dependent kinases (CDKs) is considered a promising strategy for cancer treatment due to their role in cell cycle regulation. However, CDK inhibitors with no selectivity among CDK families have not been approved. A CDK inhibitor with high selectivity for CDK4/6 exhibited significant treatment effects on breast cancer and has become a heavy bomb on the market. Subsequently, resistance gradually decreased the efficacy of selective CDK4/6 inhibitors in breast cancer treatment. In this review, we first introduce the development of selective CDK4/6 inhibitors and then explain the role of CDK2 activation in inducing resistance to CDK4/6 inhibitors. Moreover, we focused on the development of CDK2/4/6 inhibitors and selective CDK2 inhibitors, which will aid in the discovery of novel CDK inhibitors targeting the cell cycle in the future.
Collapse
Affiliation(s)
- Weijiao Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xujie Zhuang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuanyuan Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Huanaoyu Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Linhu Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Sikai Feng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
11
|
Wurlina W, Meles DK, Mustofa I, Khairullah AR, Putra DMS, Suwasanti N, Akintunde AO, Utama S, Mulyati S, Wasito W, Raissa R, Ahmad RZ, Julaeha J, Ekawasti F. Alkaloid fraction of Achyranthes aspera Linn triggers breast cancer apoptosis in mice ( Mus musculus) model. Open Vet J 2025; 15:1279-1288. [PMID: 40276193 PMCID: PMC12017709 DOI: 10.5455/ovj.2025.v15.i3.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 02/15/2025] [Indexed: 04/26/2025] Open
Abstract
Background Breast cancer affects women of various ages, and its recurrence is a significant cause of death. The search for potent anticancer compounds of herbal origin with well-defined mechanisms of action is an essential focus of current research. Aim This study aimed to investigate the effects of alkaloids in Achyranthes aspera Linn (AAL) leaf extract on necrosis, apoptosis, and related molecular markers, namely, cyclin-dependent kinase 1, Bcl-2 associated X-protein (Bax), rat sarcoma virus (Ras), cytochrome (Cyt) c, and apoptotic activating factor-1 (Apaf-1), in mice models. Methods Thirty mice with breast cancer were randomly divided into five groups. The negative control group only received distilled water daily. Mice in the positive control group (PCG) were administered methotrexate (15 mg/Kg) daily. The T1, T2, and T3 groups received oral orally at 75, 100, and 125 mg/Kg body weight daily for 30 days, respectively. On day 31, all mice were euthanized for the preparation of histological specimens of the mammary glands. The negative control group had the lowest number of apoptotic cells, Apaf-1, Cyt C, and Bax expression, and the highest number of viable cancer cells and Ras expression. Results The percentages of necrotic cells and breast cancer-expressed CDK-1 were not significantly (p > 0.05) different among groups. The percentage of apoptotic cells, Apaf-1, and Cyt c, was highest in T3. Conversely, the percentage of viable cells and breast cancer-expressing Ras was lowest in T3. Conclusion Treatment with 125 mg/Kg AAL suppressed cancer cell growth in breast cancer-bearing mice. Further research is necessary to determine the complete signaling mechanism.
Collapse
Affiliation(s)
- Wurlina Wurlina
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Dewa Ketut Meles
- Division of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Imam Mustofa
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | | | - Niluh Suwasanti
- Department of Clinical Pathology, Faculty of Medicine, Universitas Katolik Widya Mandala Surabaya, Surabaya, Indonesia
| | - Adeyinka Oye Akintunde
- Department of Agriculture and Industrial Technology, Babcock University, Ilishan Remo State, Nigeria
| | - Suzanita Utama
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sri Mulyati
- Division of Veterinary Reproduction, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Wasito Wasito
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Ricadonna Raissa
- Department of Pharmacology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Riza Zainuddin Ahmad
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Julaeha Julaeha
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Fitrine Ekawasti
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| |
Collapse
|
12
|
Shi C, Zhang F, He Q, Man J, Mu Y, Zhao J, Zhu L, Loor JJ, Luo J. ADCY5 Gene Affects Seasonal Reproduction in Dairy Goats by Regulating Ovarian Granulosa Cells Steroid Hormone Synthesis. Int J Mol Sci 2025; 26:1622. [PMID: 40004085 PMCID: PMC11855216 DOI: 10.3390/ijms26041622] [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: 12/26/2024] [Revised: 02/01/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Follicle development in dairy goats is lower after induced estrus during the non-breeding season, reducing conception rates and challenging year-round milk supply. This study investigated follicle development during the breeding and non-breeding seasons and explored molecular mechanisms for variations in the proportions of follicles of different sizes using ovarian RNA-seq and in vitro experiments. Induced estrus during the non-breeding season used a simulated breeding season short photoperiod and male effect methods, while the male effect method was used during the breeding season. This study identified an increase in follicle size during the breeding season and performed RNA-seq on ovaries to explore the underlying causes. The RNA-seq analysis elucidated pathways associated with cellular and hormonal metabolism and identified adenylyl cyclase 5 (ADCY5) as a key differentially expressed gene. In vitro experiments demonstrated that interfering with ADCY5 in ovarian granulosa cells (GCs) reduced steroid synthesis. Conversely, the overexpression of ADCY5 increased steroid synthesis. ADCY5 affects the biological function of GCs and consequently influences follicle development through the cAMP-response element binding protein (CREB) and p38 mitogen-activated protein kinase phosphorylation (MAPK) pathways. Overall, our findings demonstrate that follicle development in dairy goats differs between the breeding and non-breeding seasons and that the differential expression levels of the ADCY5 gene contribute to this discrepancy.
Collapse
Affiliation(s)
- Chenbo Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Fuhong Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Qiuya He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Jianjun Man
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Yuanpan Mu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Jianqing Zhao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Lu Zhu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| | - Juan J. Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA;
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (C.S.); (F.Z.); (Q.H.); (J.M.); (Y.M.); (J.Z.); (L.Z.)
| |
Collapse
|
13
|
Nakao Y, Okamoto K, Tazawa I, Nishijima T, Furuno N, Sakuma T, Yamamoto T, Takeuchi T, Hayashi T. Effect of Cdk1 gene disruption on cell cycle progression in newt cells. Dev Growth Differ 2025; 67:85-93. [PMID: 39776058 DOI: 10.1111/dgd.12958] [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: 10/14/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025]
Abstract
Cyclin-dependent kinases (CDKs) are key regulators of cell cycle progression, in conjunction with cyclins. The cyclin-CDK system is highly conserved among eukaryotes, and CDK1 is considered essential for progression through the M phase. However, the extent to which cell cycle progression depends on CDK1 varies between cell types. Therefore, a range of cell types must be analyzed to comprehensively elucidate the role of CDK1. Cdk1-knockout mice exhibit lethality at an early developmental stage, specifically before the differentiation of various cell types. The aim of the present study was to characterize the effects of CDK1 deficiency in amphibian newts. Cdk1 was disrupted by injecting fertilized newt eggs with CRISPR/Cas9, and the resulting effects on embryonic development and cell proliferation were then evaluated. In both wild-type and Cdk1-crispant newt embryos, CDK1 protein was either stored in the egg until late embryogenesis or potentially derived from maternal mRNA, which may also be stored during this period. The embryos survived to the hatching stage, during which the cells responsible for forming the basic organs differentiated. To further characterize the long-term effects of Cdk1 knockout, parabiosis experiments were conducted using wild-type embryos and Cdk1 crispants. The results suggested that an endocycle occurred in the crispant larvae, as evidenced by increases in the size of several types of cells. It is anticipated that studies using newts will provide further insights into the role of Cdk1 in regulating the cell cycle.
Collapse
Affiliation(s)
- Yuta Nakao
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Kazuko Okamoto
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ichiro Tazawa
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Tatsuro Nishijima
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Nobuaki Furuno
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Tetsushi Sakuma
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Takashi Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- Hiroshima University Genome Editing Center, Higashi-Hiroshima, Japan
| | - Takashi Takeuchi
- Department of Biomedical Sciences, School of Life Sciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshinori Hayashi
- Amphibian Research Center, Hiroshima University, Higashi-Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
14
|
Apoorvha JP, Brindha S, Ganesan M, Roy S. Weighted gene co-expression network analysis reveals the hub genes and molecular mechanism of quiescence. 3 Biotech 2025; 15:42. [PMID: 39829641 PMCID: PMC11735823 DOI: 10.1007/s13205-024-04203-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025] Open
Abstract
The hematopoietic stem cell (HSC) continues their functional integrity and return to quiescence quickly even after inflammatory and other proliferative stress. The mechanism which is responsible for this highly regulatory process is not understood clearly. Previous results have shown that CD53 is noticeably upregulated in HSCs in response to a variety of stimuli. Gene expression profile using RNASeq data of HSCs from the bone marrow and spleen of CD53 knock out and their wild-type littermate had been deposited by Greenberg and co-authors, in GEO database, "GSE219050". They reported that knockout of CD53 promotes continued cell cycle. To identify key genes and specific processes are affected in absence of CD53, we applied weighted gene co-expression analysis. The results show that cyan module is correlated and dark red and light cyan are anti-correlated with CD53 loss. CDK1 is identified as more connected gene or hub gene in cyan module and it is upregulated in the absence of CD53. Likewise, hub genes from dark-red module are EP300, EGF, MCL1, LPL and IGF1R. The gene enrichment analysis depicts, two biological processes, MAPK cascade and Delta Notch signalling were suppressed. Similarly, the biological processes involved in light-cyan module are chromatin organisation and hub genes are Ehmt2, Ezh2, Kdm1a, Rbbp4, Esr1 and Mysm1. It uncovers the roles of CD53 in chromatin organisation, and MAPK cascade and Delta Notch signalling are the major contributors in quiescence mechanism. These findings might provide a new avenue in quiescence research.
Collapse
Affiliation(s)
- J. P. Apoorvha
- Department of Biotechnology, Rajalakshmi Engineering College, Thandalam, Chennai, 602105 India
| | - S. Brindha
- Department of Biotechnology, Rajalakshmi Engineering College, Thandalam, Chennai, 602105 India
| | - M. Ganesan
- Department of Biotechnology, Rajalakshmi Engineering College, Thandalam, Chennai, 602105 India
| | - Sujata Roy
- Department of Biotechnology, Rajalakshmi Engineering College, Thandalam, Chennai, 602105 India
| |
Collapse
|
15
|
Wang B, Bao L, Li X, Sun G, Yang W, Xie N, Lei L, Chen W, Zhang H, Chen M, Zhao X, Wan X, Yuan R, Jiang H. Identification and validation of the important role of KIF11 in the development and progression of endometrial cancer. J Transl Med 2025; 23:48. [PMID: 39806429 PMCID: PMC11727483 DOI: 10.1186/s12967-025-06081-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Human kinesin family member 11 (KIF11) plays a vital role in regulating the cell cycle and is implicated in the tumorigenesis and progression of various cancers, but its role in endometrial cancer (EC) is still unclear. Our current research explored the prognostic value, biological function and targeting strategy of KIF11 in EC through approaches including bioinformatics, machine learning and experimental studies. METHODS The GSE17025 dataset from the GEO database was analyzed via the limma package to identify differentially expressed genes (DEGs) in EC. Functional enrichment analysis of the DEGs was conducted using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. DEGs were further screened for hub genes through protein-protein interaction (PPI) network analysis and machine learning. The role of the hub gene KIF11 in EC was analyzed using clinical data from the TCGA database. The expression of KIF11 in EC was subsequently validated in clinical samples. In vitro experiments were utilized to evaluate the effects of KIF11 on biological functions such as proliferation, migration, apoptosis, and the cell cycle in endometrial cancer cells. RESULTS A total of 877 DEGs, which are widely involved in important biological processes such as cell division, tubulin binding, and the cell cycle, were identified. Through PPI network analysis and machine learning, KIF11 was selected as the hub gene for subsequent analysis and experimental validation. An analysis of TCGA data revealed that KIF11 is highly expressed in EC and is associated with tumor grade, stage, and a low survival rate. The overexpression of KIF11 in tumor tissues was further confirmed in EC patient samples. KIF11 knockdown had inhibitory effects on cell proliferation, migration and invasion. Flow cytometry analysis revealed that KIF11 knockdown induced G2/M phase arrest and promoted apoptosis in EC cells. CONCLUSION Our study demonstrated that KIF11 was upregulated in EC and was strongly associated with a poor prognosis. Notably, we found that reduced KIF11 expression inhibited EC cell proliferation, migration and invasion. KIF11 knockdown caused more EC cells to arrest in the G2/M phase and undergo apoptosis. The findings of our study emphasized that KIF11 may be a promising prognostic biomarker and therapeutic target for EC patients.
Collapse
Affiliation(s)
- Biying Wang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Lunmin Bao
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Xiaoduo Li
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Guang Sun
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Wu Yang
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Nanzi Xie
- Department of Pathology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Ling Lei
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Wei Chen
- Department of Pathology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Hailong Zhang
- Department of Laboratory Medicine, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Man Chen
- Department of Gynaecology, Peoples' Hospital of Anshun City, Guizhou, 561000, Anshun, China
| | - Xing Zhao
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
| | - Xiufang Wan
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Rui Yuan
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China
| | - Hongmei Jiang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou, 550000, China.
- School of Basic Medicine, Guizhou Medical University, Guizhou, 550000, Guiyang, China.
- Guizhou Nursing Vocational College, Guizhou, 550000, Guiyang, China.
| |
Collapse
|
16
|
Hu HT, Wang UTT, Chen BC, Hsueh YP, Wang TF. Ki-67 and CDK1 control the dynamic association of nuclear lipids with mitotic chromosomes. J Lipid Res 2025; 66:100731. [PMID: 39706365 PMCID: PMC11786767 DOI: 10.1016/j.jlr.2024.100731] [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: 10/15/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024] Open
Abstract
Nuclear lipids play roles in regulatory processes, such as signaling, transcriptional regulation, and DNA repair. In this report, we demonstrate that nuclear lipids may contribute to Ki-67-regulated chromosome integrity during mitosis. In COS-7 cells, nuclear lipids are enriched at the perichromosomal layer and excluded from intrachromosomal regions during early mitosis but are then detected in intrachromosomal regions during late mitosis, as revealed by TT-ExM (expansion microscopy with trypsin digestion and tyramide signal amplification), an improved expansion microscopy technique that enables high-sensitivity and super-resolution imaging of proteins, lipids, and nuclear DNA. The nuclear nonhistone protein Ki-67 acts as a surfactant to form a repulsive molecular brush around fully condensed sister chromatids in early mitosis, preventing the diffusion or penetration of nuclear lipids into intrachromosomal regions. Ki-67 is phosphorylated during mitosis by cyclin-dependent kinase 1 (CDK1), the best-known master regulator of the cell cycle. Both Ki-67 knockdown and reduced Ki-67 phosphorylation by CDK1 inhibitors allow nuclear lipids to penetrate chromosomal regions. Thus, both Ki-67 protein level and phosphorylation status during mitosis appear to influence the perichromosomal distribution of nuclear lipids. Ki-67 knockdown and CDK1 inhibition also lead to uneven chromosome disjunction between daughter cells, highlighting the critical role of this regulatory mechanism in ensuring accurate chromosome segregation. Given that Ki-67 has been proposed to promote chromosome individualization and establish chromosome-cytoplasmic compartmentalization during open mitosis in vertebrates, our results reveal that nuclear lipid enrichment at the perichromosomal layer enhances the ability of Ki-67 to form a protective perichromosomal barrier (chromosome envelope), which is critical for correct chromosome segregation and maintenance of genome integrity during mitosis.
Collapse
Affiliation(s)
- Hsiao-Tang Hu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ueh-Ting Tim Wang
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan; Department of Photonics, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Bi-Chang Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| | - Ting-Fang Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
17
|
Zhang Y, Li R, Li H, Huang Y, Mei Y, Zheng Y, Guo Y, Zhou Z, Yong Z, Zhao Y, Dong W, Gao J, Cao X. Morpho-histological and Transcriptome Analysis Reveal the Unreduced Sperm Formation Mechanism in cdk1-Depletion Zebrafish. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:1206-1218. [PMID: 39243300 DOI: 10.1007/s10126-024-10366-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024]
Abstract
Cyclin-dependent kinases (Cdks) are major molecules related to cell cycle regulation. Polyploidy can be caused by the production of unreduced gametes, which is often related to the abnormal cell cycle of germ cells. Here, we successfully constructed a cdk1 mutation line (cdk1+/-) in zebrafish, a commonly used model organism. It showed that cdk1 depletion resulted in the generation of both polyploid and aneuploid embryos of WT♀ × cdk1+/-♂ zebrafish. In addition to normal sperms (1N), the depletion of cdk1 in zebrafish also led to the production of some large-head 2N sperms and higher ploidy sperms. Results of bivalent analysis of testis and ultrastructure analysis of spermatogonia suggested that the production of these large-head sperms was due to spermatogonia chromosome doubling in cdk1+/- zebrafish. Transcriptome analysis revealed aberrant expressions of some cell cycle and DNA replication-related genes in the early testis of cdk1+/- zebrafish relative to WT zebrafish. Through STRING correlation analysis, we further proved that cdk1 depletion affected the mitosis process and endoduplication initiation of spermatogonia by regulating expressions of some proteins related to cell cycle (i.e., Espl1 and Pp1) and DNA replication (i.e., Orc1 and Rnaseh2b), thereby leading to the formation of unreduced sperms. This study provides important information on revealing the molecular mechanisms of unreduced gamete formation caused by cdk1 mutation. Meanwhile, it also provides an important reference for the creation of fish polyploid germplasm.
Collapse
Affiliation(s)
- Yunbang Zhang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, No. 1 Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Rongyun Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hui Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuwei Huang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yihui Mei
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuxuan Zheng
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yankun Guo
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zihao Zhou
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhonglin Yong
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ying Zhao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjing Dong
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, No. 1 Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Huazhong Agricultural University, Wuhan, 430070, China.
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, No. 1 Shizishan Street, Hongshan District, Wuhan, 430070, Hubei Province, People's Republic of China.
| |
Collapse
|
18
|
Chadha Y, Khurana A, Schmoller KM. Eukaryotic cell size regulation and its implications for cellular function and dysfunction. Physiol Rev 2024; 104:1679-1717. [PMID: 38900644 PMCID: PMC11495193 DOI: 10.1152/physrev.00046.2023] [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: 12/26/2023] [Revised: 05/24/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Depending on cell type, environmental inputs, and disease, the cells in the human body can have widely different sizes. In recent years, it has become clear that cell size is a major regulator of cell function. However, we are only beginning to understand how the optimization of cell function determines a given cell's optimal size. Here, we review currently known size control strategies of eukaryotic cells and the intricate link of cell size to intracellular biomolecular scaling, organelle homeostasis, and cell cycle progression. We detail the cell size-dependent regulation of early development and the impact of cell size on cell differentiation. Given the importance of cell size for normal cellular physiology, cell size control must account for changing environmental conditions. We describe how cells sense environmental stimuli, such as nutrient availability, and accordingly adapt their size by regulating cell growth and cell cycle progression. Moreover, we discuss the correlation of pathological states with misregulation of cell size and how for a long time this was considered a downstream consequence of cellular dysfunction. We review newer studies that reveal a reversed causality, with misregulated cell size leading to pathophysiological phenotypes such as senescence and aging. In summary, we highlight the important roles of cell size in cellular function and dysfunction, which could have major implications for both diagnostics and treatment in the clinic.
Collapse
Affiliation(s)
- Yagya Chadha
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Arohi Khurana
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Kurt M Schmoller
- Institute of Functional Epigenetics, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
19
|
Teotia V, Jha P, Chopra M. Discovery of Potential Inhibitors of CDK1 by Integrating Pharmacophore-Based Virtual Screening, Molecular Docking, Molecular Dynamics Simulation Studies, and Evaluation of Their Inhibitory Activity. ACS OMEGA 2024; 9:39873-39892. [PMID: 39346877 PMCID: PMC11425824 DOI: 10.1021/acsomega.4c05414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/01/2024]
Abstract
The ability of CDK1 to compensate for the absence of other cell cycle CDKs poses a great challenge to treat cancers that overexpress these proteins. Despite several studies focusing on the area, there are no FDA-approved drugs selectively targeting CDK1. Here, the study aimed to develop potential CDK1 selective inhibitors through drug repurposing and leveraging the structural insights provided by the hit molecules generated. Approximately 280,000 compounds from DrugBank, Selleckchem, Otava and an in-house library were screened initially based on fit values using 3D QSAR pharmacophores built for CDK1 and subsequently through Lipinski, ADMET, and TOPKAT filters. 10,310 hits were investigated for docking into the binding site of CDK1 determined using the crystal structure of human CDK1 in complex with NU6102. The best 55 hits with better docking scores were further analyzed, and 12 hits were selected for 100 ns MD simulations followed by binding energy calculations using the MM-PBSA method. Finally, 10 hit molecules were tested in an in vitro CDK1 Kinase inhibition assay. Out of these, 3 hits showed significant CDK1 inhibitory potential with IC50 < 5 μM. These results indicate these compounds can be used to develop subtype-selective CDK1 inhibitors with better efficacy and reduced toxicities in the future.
Collapse
Affiliation(s)
- Vineeta Teotia
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Prakash Jha
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| | - Madhu Chopra
- Laboratory
of Molecular Modeling and Anti-Cancer Drug Development, Dr. B. R.
Ambedkar Center for Biomedical Research, University of Delhi, Delhi 110007, India
| |
Collapse
|
20
|
Paul Chowdhuri S, Das BB. TDP1 phosphorylation by CDK1 in mitosis promotes MUS81-dependent repair of trapped Top1-DNA covalent complexes. EMBO J 2024; 43:3710-3732. [PMID: 39014228 PMCID: PMC11377750 DOI: 10.1038/s44318-024-00169-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/18/2024] Open
Abstract
Topoisomerase 1 (Top1) controls DNA topology, relieves DNA supercoiling during replication and transcription, and is critical for mitotic progression to the G1 phase. Tyrosyl-DNA phosphodiesterase 1 (TDP1) mediates the removal of trapped Top1-DNA covalent complexes (Top1cc). Here, we identify CDK1-dependent phosphorylation of TDP1 at residue S61 during mitosis. A TDP1 variant defective for S61 phosphorylation (TDP1-S61A) is trapped on the mitotic chromosomes, triggering DNA damage and mitotic defects. Moreover, we show that Top1cc repair in mitosis occurs via a MUS81-dependent DNA repair mechanism. Replication stress induced by camptothecin or aphidicolin leads to TDP1-S61A enrichment at common fragile sites, which over-stimulates MUS81-dependent chromatid breaks, anaphase bridges, and micronuclei, ultimately culminating in the formation of 53BP1 nuclear bodies during G1 phase. Our findings provide new insights into the cell cycle-dependent regulation of TDP1 dynamics for the repair of trapped Top1-DNA covalent complexes during mitosis that prevents genomic instability following replication stress.
Collapse
Affiliation(s)
- Srijita Paul Chowdhuri
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Benu Brata Das
- Laboratory of Molecular Biology, School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & B, Raja S. C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
21
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. PLoS Genet 2024; 20:e1011387. [PMID: 39226333 PMCID: PMC11398662 DOI: 10.1371/journal.pgen.1011387] [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: 04/02/2024] [Revised: 09/13/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogaster wing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Lauren L Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| | - Brian R Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, Indiana, United States of America
| |
Collapse
|
22
|
Nazari MHD, Heidarian R, Masoudnia M, Dastjerdi RA, Talkhounche PG, Taleahmad S. Targeting GLI1 and BAX by nanonoscapine could impede prostate adenocarcinoma progression. Sci Rep 2024; 14:18977. [PMID: 39152150 PMCID: PMC11329793 DOI: 10.1038/s41598-024-65968-4] [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: 12/15/2023] [Accepted: 06/25/2024] [Indexed: 08/19/2024] Open
Abstract
Prostate cancer as a critical global health issue, requires the exploration of a novel therapeutic approach. Noscapine, an opium-derived phthalide isoquinoline alkaloid, has shown promise in cancer treatment thanks to its anti-tumorigenic properties. However, limitations such as low bioavailability and potential side effects have hindered its clinical application. This study introduces nanonoscapine as a novel medication to overcome these challenges, leveraging the advantages of improved drug delivery and efficacy achieved in nanotechnology. We monitored the effects of nanonoscapine on the androgen-sensitive human prostate adenocarcinoma cell line, LNCaP, investigating its impact on GLI1 and BAX genes' expressions, crucial regulators of cell cycle and apoptosis. Our findings, from MTT assays, flow cytometry, and gene expression analyses, have demonstrated that nanonoscapine effectively inhibits prostate cancer cell proliferation by inducing G2/M phase arrest and apoptosis. Furthermore, through bioinformatics and computational analyses, we have revealed the underlying molecular mechanisms, underscoring the therapeutic potential of nanonoscapine in enhancing patient outcomes. This study highlights the significance of nanonoscapine as an alternative or adjunct treatment to conventional chemotherapy, warranting further investigation in clinical settings.
Collapse
Affiliation(s)
- Mohammad Hossein Derakhshan Nazari
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Ronak Heidarian
- Department of Developmental Biology, Kharazmi University, Tehran, Iran
| | - Mina Masoudnia
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rana Askari Dastjerdi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Parnian Ghaedi Talkhounche
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
23
|
Tao S, Long X, Gong P, Yu X, Tian L. Phosphoproteomics Reveals Novel Insights into the Pathogenesis and Identifies New Therapeutic Kinase Targets of Ulcerative Colitis. Inflamm Bowel Dis 2024; 30:1367-1378. [PMID: 38085663 DOI: 10.1093/ibd/izad291] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Indexed: 08/02/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic recurrent inflammatory disease with unclear etiology. Currently, safe and effective treatment options for UC remain to be developed. Kinases, which catalyze the phosphorylation of substrates, have emerged as promising therapeutic targets for inflammatory diseases. We clarified the kinase activity profile and phosphorylation network in UC and aimed to reveal new pathogenic mechanisms and potential therapeutic targets. METHODS We first performed the phosphoproteomic analysis of rectal tissues from UC patients and healthy individuals. Further bioinformatic analyses revealed the remodeling of key kinases and signaling pathways. Then, we conducted a screening of kinases to identify new potential therapeutic targets through in vivo and in vitro experiments. RESULTS Phosphoproteomics revealed a drastic remodeling of signaling pathways in UC, such as pathways related to tight junction, adhesion junction, and necroptosis. Additionally, the activity of kinases such as CDK2, CLK1 and AURKB were significantly changed. Additional screening of these kinases identified CDK2 as a potential therapeutic target for UC, as inhibiting CDK2 effectively alleviated dextran sulfate sodium-induced colitis in mice. Further research revealed that suppressing CDK2 remarkably inhibited RIPK1, RIPK3, and MLKL phosphorylation, as well as MLKL oligomerization, thereby inhibiting epithelial necroptosis and protecting the intestinal barrier. CONCLUSIONS Our research deepened the understanding of UC pathogenesis through the lens of phosphorylation. Moreover, we identified CDK2 as a new potential therapeutic target for UC, revealing a novel role for CDK2 in necroptosis.
Collapse
Affiliation(s)
- Sifan Tao
- Department of Gastroenterology, The Third Xiangya Hospital, The Central South University, Changsha, China
- Key Laboratory of Non-resolving Inflammation and Cancer of the Hunan Province, The Third Xiangya Hospital, The Central South University, Changsha, China
| | - Xiuyan Long
- Department of Gastroenterology, The Third Xiangya Hospital, The Central South University, Changsha, China
| | - Pan Gong
- Department of Gastroenterology, The Third Xiangya Hospital, The Central South University, Changsha, China
| | - Xiaoyu Yu
- Department of Gastroenterology, The Third Xiangya Hospital, The Central South University, Changsha, China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital, The Central South University, Changsha, China
| |
Collapse
|
24
|
Yin K, Büttner M, Deligiannis IK, Strzelecki M, Zhang L, Talavera-López C, Theis F, Odom DT, Martinez-Jimenez CP. Polyploidisation pleiotropically buffers ageing in hepatocytes. J Hepatol 2024; 81:289-302. [PMID: 38583492 DOI: 10.1016/j.jhep.2024.03.043] [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: 05/05/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND & AIMS Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS We performed single-nucleus RNA sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent strains that were haploinsufficient for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS We observed that ageing in wild-type mice results in nuclei polyploidy and a marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in the CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A-haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbations. CONCLUSIONS Our results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome. IMPACT AND IMPLICATIONS The functional role of hepatocyte polyploidisation during ageing is poorly understood. Using single-nucleus RNA sequencing and BaseScope approaches, we have studied ploidy dynamics during ageing in murine livers with non-deleterious genetic perturbations. We have identified that hepatocytes present different cellular states and the ability to buffer ageing-associated dysfunctions. Tetraploid nuclei exhibit robust transcriptional networks and are better adapted to genomically overcome perturbations. Novel therapeutic interventions aimed at attenuating age-related changes in tissue function could be exploited by manipulation of ploidy dynamics during chronic liver conditions.
Collapse
Affiliation(s)
- Kelvin Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany
| | | | | | - Liwei Zhang
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Carlos Talavera-López
- Division of Infectious Diseases and Tropical Medicine, Ludwig-Maximilian-Universität Klinikum, Germany
| | - Fabian Theis
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching. Munich, Germany; German Cancer Research Centre, Heidelberg, Germany.
| | - Duncan T Odom
- German Cancer Research Center, Division of Regulatory Genomics and Cancer Evolution (B270), Heidelberg, Germany; Cancer Research UK Cambridge Institute, University of Cambridge, CB20RE, United Kingdom.
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany; Institute of Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
25
|
Ingebriktsen LM, Svanøe AA, Myrmel Sæle AK, Humlevik ROC, Toska K, Kalvenes MB, Aas T, Heie A, Askeland C, Knutsvik G, Stefansson IM, Akslen LA, Hoivik EA, Wik E. Age-Related Clusters and Favorable Immune Phenotypes in Young Breast Cancer Patients. Mod Pathol 2024; 37:100529. [PMID: 38810731 DOI: 10.1016/j.modpat.2024.100529] [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: 11/03/2023] [Revised: 05/12/2024] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
Breast cancer (BC) patients aged <40 years at diagnosis experience aggressive disease and poorer survival compared with women diagnosed with BC at 40 to 49 years, but the age-related biology is described to little extent. Here, we explored transcriptional alterations in BC to gain better understanding of age-related tumor biology. We studied a subset of the Bergen in-house cohort (n = 127; age range, 26-49 years) and used the NanoString Breast Cancer 360 expression panel on formalin-fixed paraffin-embedded BC tissue, and publicly available global BC messenger RNA expression data (n = 204; age range, 22-49 years), to explore differentially expressed genes between the young (age <40 years) and older (age 40-49 years) patients. Unsupervised hierarchical clustering was applied to identify gene expression-based patient clusters. We applied established computational approaches to define the PAM50 subtypes, risk of recurrence scores (ROR), and risk groups and to infer the proportions of 22 immune cell types from bulk gene expression profiles of patients aged <50 years at BC diagnosis. Differentially expressed genes and gene sets were investigated using OncoEnrichR and g:Profiler to describe functional profiles and pathway enrichment. We identified 4 age-related patient clusters presenting distinct characteristics of PAM50 subtypes and ROR profiles, which demonstrated independent prognostic value when adjusted for traditional clinicopathologic variables and the known molecular subtypes. Our findings showed better survival than expected in the basal-enriched cluster 2 and in triple-negative and basal-like BC. Deconvolution analyses of immunophenotypes indicated higher levels of M0 and M1 macrophages than M2 macrophages in subsets of young BC. Our approach identifies age-based patient clusters with distinct clinicopathologic profiles, to a large extent overlapping with the PAM50 subtypes, although with independent prognostic values in multivariate survival analyses. The patient clusters provided new insight in the immune cell distribution across tumor subtypes, potentially contributing to survival differences between the clusters and the molecular subtypes and indicating age-related mechanisms improving outcome. Our study confirms the applicability of ROR as a valid prognosticator also in a young BC cohort.
Collapse
Affiliation(s)
- Lise Martine Ingebriktsen
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway
| | - Amalie Abrahamsen Svanøe
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway
| | - Anna Kristine Myrmel Sæle
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Rasmus Olai Collett Humlevik
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway
| | - Karen Toska
- Section for Cancer Genomics, Haukeland University Hospital, Bergen, Norway
| | - May Britt Kalvenes
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway
| | - Turid Aas
- Department of Surgery, Section for Breast and Endocrine Surgery, Haukeland University Hospital, Bergen, Norway
| | - Anette Heie
- Department of Surgery, Section for Breast and Endocrine Surgery, Haukeland University Hospital, Bergen, Norway
| | - Cecilie Askeland
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Gøril Knutsvik
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Ingunn Marie Stefansson
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Lars Andreas Akslen
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Erling Andre Hoivik
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Wik
- Centre for Cancer Biomarkers CCBIO, Section for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
26
|
Wang C, Shan S, Li X, Wang H, Qi J, Zhao S. The role of GADD45G methylation in endometrial cancer: Insights into CDK1/CCNB1 activation and therapeutic opportunities. J Cancer Res Ther 2024; 20:1214-1223. [PMID: 39206984 DOI: 10.4103/jcrt.jcrt_2103_23] [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: 09/17/2023] [Accepted: 03/01/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Accumulating evidence suggests the significant involvement of GADD45G in the development of various cancers. This study investigates GADD45G's involvement and methylation status in endometrial cancer (EC), along with molecular mechanisms and potential therapies. METHODS The expression of GADD45G in EC tissues and controls was evaluated using RNA-seq, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting (WB). Methylation-specific PCR (MSP) evaluated GADD45G's methylation status. Protein-protein interaction (PPI) prediction identified potential interactors of GADD45G, and co-immunoprecipitation (co-IP) confirmed GADD45G interact with Cyclin-dependent kinase 1 (CDK1) and cyclin B1 (CCNB1). Several cell behavior assays were conducted in both in vitro and in vivo settings to comprehensively understand the impact of GADD45G dysregulation in EC. RESULTS Our findings revealed a significant decrease in the expression of GADD45G in endometrial cancer tissues and cells, which was attributed to its methylation status. Reduced GADD45G expression correlated with increased invasive behaviors in EC cells. Furthermore, GADD45G negatively regulated CDK1 and CCNB1, promoting invasive behaviors at transcript and protein levels. CONCLUSION This study demonstrated that the downregulation of GADD45G, mediated by methylation, facilitates the invasive behaviors of EC cells through interaction with the CDK1/CCNB1. These findings enhance understanding of the molecular mechanisms underlying endometrial cancer and suggest potential therapeutic strategies targeting GADD45G for treatment.
Collapse
Affiliation(s)
- Chunxiao Wang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Gynecology, Cangzhou People's Hospital, Cangzhou, Hebei, China
| | - Shuzhi Shan
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinjun Li
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Huifang Wang
- Department of Gynecology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Jie Qi
- Department of Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Sufen Zhao
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
27
|
Farhangniya M, Samadikuchaksaraei A, Mohamadi Farsani F. Exploring Co-expression Modules-Traits Correlation through Weighted Gene Co-expression Network Analysis: A Promising Approach in Wound Healing Research. Med J Islam Repub Iran 2024; 38:82. [PMID: 39678778 PMCID: PMC11644100 DOI: 10.47176/mjiri.38.82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Indexed: 12/17/2024] Open
Abstract
Background The skin is the biggest organ in the body and has several important functions in protection and regulation. However, wound development can disrupt the natural healing process, leading to challenges such as chronic wounds, persistent infections, and impaired angiogenesis. These issues not only affect individuals' well-being but also pose significant economic burdens on healthcare systems. Despite advancements in wound care research, managing chronic wounds remains a pressing concern, with obstacles such as persistent infection and impaired angiogenesis hindering the healing process. Understanding the complex genetic pathways involved in wound healing is crucial for developing effective therapeutic strategies and reducing the socio-economic impact of chronic wounds. Weighted Gene Co-Expression Network Analysis (WGCNA) offers a promising approach to uncovering key genes and modules associated with different stages of wound healing, providing valuable insights for targeted interventions to enhance tissue repair and promote efficient wound healing. Methods Data collection involved retrieving microarray gene expression datasets from the Gene Expression Omnibus website, with 65 series selected according to inclusion and exclusion criteria. Preprocessing of raw data was performed using the Robust MultiArray Averaging approach for background correction, normalization, and gene expression calculation. Weighted Gene Co-Expression Network Analysis was employed to identify co-expression patterns among genes associated with wound healing processes. This involved steps such as network construction, topological analysis, module identification, and association with clinical traits. Functional analysis included enrichment analysis and identification of hub genes through gene-gene functional interaction network analysis using the GeneMANIA database. Results The analysis using WGCNA indicated significant correlations between wound healing and the black, brown, and light green modules. These modules were further examined for their relevance to wound healing traits and subjected to functional enrichment analysis. A total of 16 genes were singled out as potential hub genes critical for wound healing. These hub genes were then scrutinized, revealing a gene-gene functional interaction network within the module network based on the KEGG enrichment database. Noteworthy pathways such as MAPK, EGFR, and ErbB signaling pathways, as well as essential cellular processes including autophagy and mitophagy, emerged as the most notable significant pathways. Conclusion We identified consensus modules relating to wound healing across nine microarray datasets. Among these, 16 hub genes were uncovered within the brown and black modules. KEGG enrichment analysis identified co-expression genes within these modules and highlighted pathways most closely associated with the development of wound healing traits, including autophagy and mitophagy. The hub genes identified in this study represent potential candidates for future research endeavors. These findings serve as a stepping stone toward further exploration of the implications of these co-expressed modules on wound healing traits.
Collapse
Affiliation(s)
- Mansoureh Farhangniya
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Health Metrics Research Center, Iranian Institute for Health Sciences Research, ACECR, Tehran, Iran
| | - Ali Samadikuchaksaraei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
28
|
Ma Z, Li DX, Lan X, Bubelenyi A, Vyhlidal M, Kunze M, Sommerfeldt M, Adesida AB. Short-term response of primary human meniscus cells to simulated microgravity. Cell Commun Signal 2024; 22:342. [PMID: 38907358 PMCID: PMC11191296 DOI: 10.1186/s12964-024-01684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/27/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Mechanical unloading of the knee articular cartilage results in cartilage matrix atrophy, signifying the osteoarthritic-inductive potential of mechanical unloading. In contrast, mechanical loading stimulates cartilage matrix production. However, little is known about the response of meniscal fibrocartilage, a major mechanical load-bearing tissue of the knee joint, and its functional matrix-forming fibrochondrocytes to mechanical unloading events. METHODS In this study, primary meniscus fibrochondrocytes isolated from the inner avascular region of human menisci from both male and female donors were seeded into porous collagen scaffolds to generate 3D meniscus models. These models were subjected to both normal gravity and mechanical unloading via simulated microgravity (SMG) for 7 days, with samples collected at various time points during the culture. RESULTS RNA sequencing unveiled significant transcriptome changes during the 7-day SMG culture, including the notable upregulation of key osteoarthritis markers such as COL10A1, MMP13, and SPP1, along with pathways related to inflammation and calcification. Crucially, sex-specific variations in transcriptional responses were observed. Meniscus models derived from female donors exhibited heightened cell proliferation activities, with the JUN protein involved in several potentially osteoarthritis-related signaling pathways. In contrast, meniscus models from male donors primarily regulated extracellular matrix components and matrix remodeling enzymes. CONCLUSION These findings advance our understanding of sex disparities in knee osteoarthritis by developing a novel in vitro model using cell-seeded meniscus constructs and simulated microgravity, revealing significant sex-specific molecular mechanisms and therapeutic targets.
Collapse
Affiliation(s)
- Zhiyao Ma
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - David Xinzheyang Li
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
- Department of Civil and Environmental Engineering, Faculty of Engineering, AB, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Adam Bubelenyi
- Faculty of Science, AB, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Margaret Vyhlidal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Mark Sommerfeldt
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
- Department of Mechanical Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, T6G 2R3, Canada.
| |
Collapse
|
29
|
Hu R, Fan W, Li S, Zhang G, Zang L, Qin L, Li R, Chen R, Zhang L, Gu W, Zhang Y, Rajagopalan S, Sun Q, Liu C. PM 2.5-induced cellular senescence drives brown adipose tissue impairment in middle-aged mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116423. [PMID: 38705039 PMCID: PMC12070286 DOI: 10.1016/j.ecoenv.2024.116423] [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] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/07/2024]
Abstract
Airborne fine particulate matter (PM2.5) exposure is closely associated with metabolic disturbance, in which brown adipose tissue (BAT) is one of the main contributing organs. However, knowledge of the phenotype and mechanism of PM2.5 exposure-impaired BAT is quite limited. In the study, male C57BL/6 mice at three different life phases (young, adult, and middle-aged) were simultaneously exposed to concentrated ambient PM2.5 or filtered air for 8 weeks using a whole-body inhalational exposure system. H&E staining and high-resolution respirometry were used to assess the size of adipocytes and mitochondrial function. Transcriptomics was performed to determine the differentially expressed genes in BAT. Quantitative RT-PCR, immunohistochemistry staining, and immunoblots were performed to verify the transcriptomics and explore the mechanism for BAT mitochondrial dysfunction. Firstly, PM2.5 exposure caused altered BAT morphology and mitochondrial dysfunction in middle-aged but not young or adult mice. Furthermore, PM2.5 exposure increased cellular senescence in BAT of middle-aged mice, accompanied by cell cycle arrest, impaired DNA replication, and inhibited AKT signaling pathway. Moreover, PM2.5 exposure disrupted apoptosis and autophagy homeostasis in BAT of middle-aged mice. Therefore, BAT in middle-aged mice was more vulnerable to PM2.5 exposure, and the cellular senescence-initiated apoptosis, autophagy, and mitochondrial dysfunction may be the mechanism of PM2.5 exposure-induced BAT impairment.
Collapse
Affiliation(s)
- Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Wenjun Fan
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Sanduo Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Li Qin
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Rucheng Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Weijia Gu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Yunhui Zhang
- Key Laboratory of Public Health Safety, Ministry of Education, School of Public Health, Fudan University, Shanghai 200433, China
| | - Sanjay Rajagopalan
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou 310053, China.
| |
Collapse
|
30
|
Mariani JN, Mansky B, Madsen PM, Salinas D, Kesmen D, Huynh NPT, Kuypers NJ, Kesel ER, Bates J, Payne C, Chandler-Militello D, Benraiss A, Goldman SA. Repression of developmental transcription factor networks triggers aging-associated gene expression in human glial progenitor cells. Nat Commun 2024; 15:3873. [PMID: 38719882 PMCID: PMC11079006 DOI: 10.1038/s41467-024-48118-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.
Collapse
Affiliation(s)
- John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Benjamin Mansky
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Pernille M Madsen
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark
| | - Dennis Salinas
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Deniz Kesmen
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Nguyen P T Huynh
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark
| | - Nicholas J Kuypers
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Erin R Kesel
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Janna Bates
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Casey Payne
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen, 2200, Denmark.
| |
Collapse
|
31
|
Xu J, Guo P, Hao S, Shangguan S, Shi Q, Volpe G, Huang K, Zuo J, An J, Yuan Y, Cheng M, Deng Q, Zhang X, Lai G, Nan H, Wu B, Shentu X, Wu L, Wei X, Jiang Y, Huang X, Pan F, Song Y, Li R, Wang Z, Liu C, Liu S, Li Y, Yang T, Xu Z, Du W, Li L, Ahmed T, You K, Dai Z, Li L, Qin B, Li Y, Lai L, Qin D, Chen J, Fan R, Li Y, Hou J, Ott M, Sharma AD, Cantz T, Schambach A, Kristiansen K, Hutchins AP, Göttgens B, Maxwell PH, Hui L, Xu X, Liu L, Chen A, Lai Y, Esteban MA. A spatiotemporal atlas of mouse liver homeostasis and regeneration. Nat Genet 2024; 56:953-969. [PMID: 38627598 DOI: 10.1038/s41588-024-01709-7] [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: 01/08/2023] [Accepted: 03/06/2024] [Indexed: 05/09/2024]
Abstract
The mechanism by which mammalian liver cell responses are coordinated during tissue homeostasis and perturbation is poorly understood, representing a major obstacle in our understanding of many diseases. This knowledge gap is caused by the difficulty involved with studying multiple cell types in different states and locations, particularly when these are transient. We have combined Stereo-seq (spatiotemporal enhanced resolution omics-sequencing) with single-cell transcriptomic profiling of 473,290 cells to generate a high-definition spatiotemporal atlas of mouse liver homeostasis and regeneration at the whole-lobe scale. Our integrative study dissects in detail the molecular gradients controlling liver cell function, systematically defining how gene networks are dynamically modulated through intercellular communication to promote regeneration. Among other important regulators, we identified the transcriptional cofactor TBL1XR1 as a rheostat linking inflammation to Wnt/β-catenin signaling for facilitating hepatocyte proliferation. Our data and analytical pipelines lay the foundation for future high-definition tissue-scale atlases of organ physiology and malfunction.
Collapse
Affiliation(s)
- Jiangshan Xu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Pengcheng Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
| | - Shijie Hao
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuncheng Shangguan
- BGI Research, Shenzhen, China
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
| | - Quan Shi
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Giacomo Volpe
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Keke Huang
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jing Zuo
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Juan An
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yue Yuan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Mengnan Cheng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Qiuting Deng
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guangyao Lai
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health and Guangzhou Medical University, Guangzhou, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Haitao Nan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Baihua Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xinyi Shentu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Liang Wu
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiaoyu Wei
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yujia Jiang
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Xin Huang
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fengyu Pan
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Yumo Song
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Ronghai Li
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Zhifeng Wang
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | - Chuanyu Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
| | - Shiping Liu
- BGI Research, Hangzhou, China
- BGI Research, Shenzhen, China
| | | | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Zhicheng Xu
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Wensi Du
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Ling Li
- China National GeneBank, BGI Research, Shenzhen, China
- Guangdong Provincial Genomics Data Center, BGI Research, Shenzhen, China
| | - Tanveer Ahmed
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Kai You
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zhen Dai
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Li Li
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Baoming Qin
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yinxiong Li
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Liangxue Lai
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dajiang Qin
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junling Chen
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Rong Fan
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Yongyin Li
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Jinlin Hou
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangzhou, China
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tobias Cantz
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Axel Schambach
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Andrew P Hutchins
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Berthold Göttgens
- Department of Haematology and Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Patrick H Maxwell
- Cambridge Institute for Medical Research, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Xun Xu
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China.
| | - Longqi Liu
- BGI Research, Hangzhou, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Ao Chen
- BGI Research, Shenzhen, China.
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
- BGI Research, Chongqing, China.
- JFL-BGI STOmics Center, BGI-Shenzhen, Chongqing, China.
| | - Yiwei Lai
- BGI Research, Hangzhou, China.
- BGI Research, Shenzhen, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
- BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China.
| | - Miguel A Esteban
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China.
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
- The Fifth Affiliated Hospital of Guangzhou Medical University-BGI Research Center for Integrative Biology, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
32
|
Fomo KN, Perumal N, Manicam C, Pfeiffer N, Grus FH. Neuroretinal Cell Culture Model as a Tool for the Development of New Therapeutic Approaches for Oxidative Stress-Induced Ocular Diseases, with a Focus on Glaucoma. Cells 2024; 13:775. [PMID: 38727311 PMCID: PMC11083839 DOI: 10.3390/cells13090775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Glaucoma is a heterogeneous group of optic neuropathies characterized by a progressive degeneration of the retinal ganglion cells (RGCs), leading to irreversible vision loss. Nowadays, the traditional therapeutic approach to glaucoma consists of lowering the intraocular pressure (IOP), which does not address the neurodegenerative features of the disease. Besides animal models of glaucoma, there is a considerable need for in vitro experimental models to propose new therapeutic strategies for this ocular disease. In this study, we elucidated the pathological mechanisms leading to neuroretinal R28 cell death after exposure to glutamate and hydrogen peroxide (H2O2) in order to develop new therapeutic approaches for oxidative stress-induced retinal diseases, including glaucoma. We were able to show that glutamate and H2O2 can induce a decrease in R28 cell viability in a concentration-dependent manner. A cell viability of about 42% was found after exposure to 3 mM of glutamate and about 56% after exposure to 100 µM of H2O2 (n = 4). Label-free quantitative mass spectrometry analysis revealed differential alterations of 193 and 311 proteins in R28 cells exposed to 3 mM of glutamate and 100 µM of H2O2, respectively (FDR < 1%; p < 0.05). Bioinformatics analysis indicated that the protein changes were associated with the dysregulation of signaling pathways, which was similar to those observed in glaucoma. Thus, the proteomic alteration induced by glutamate was associated with the inhibition of the PI3K/AKT signaling pathway. On the other hand, H2O2-induced toxicity in R28 cells was linked to the activation of apoptosis signaling and the inhibition of the mTOR and ERK/MAPK signaling pathways. Furthermore, the data show a similarity in the inhibition of the EIF2 and AMPK signaling pathways and the activation of the sumoylation and WNT/β-catenin signaling pathways in both groups. Our findings suggest that the exposure of R28 cells to glutamate and H2O2 could induce glaucoma-like neurodegenerative features and potentially provide a suitable tool for the development of new therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
| | | | | | | | - Franz H. Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Centre of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (K.N.F.); (N.P.); (C.M.); (N.P.)
| |
Collapse
|
33
|
Orhan E, Velazquez C, Tabet I, Fenou L, Rodier G, Orsetti B, Jacot W, Sardet C, Theillet C. CDK inhibition results in pharmacologic BRCAness increasing sensitivity to olaparib in BRCA1-WT and olaparib resistant in Triple Negative Breast Cancer. Cancer Lett 2024; 589:216820. [PMID: 38574883 DOI: 10.1016/j.canlet.2024.216820] [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: 10/06/2023] [Revised: 02/19/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024]
Abstract
One in three Triple Negative Breast Cancer (TNBC) is Homologous Recombination Deficient (HRD) and susceptible to respond to PARP inhibitor (PARPi), however, resistance resulting from functional HR restoration is frequent. Thus, pharmacologic approaches that induce HRD are of interest. We investigated the effectiveness of CDK-inhibition to induce HRD and increase PARPi sensitivity of TNBC cell lines and PDX models. Two CDK-inhibitors (CDKi), the broad range dinaciclib and the CDK12-specific SR-4835, strongly reduced the expression of key HR genes and impaired HR functionality, as illustrated by BRCA1 and RAD51 nuclear foci obliteration. Consequently, both CDKis showed synergism with olaparib, as well as with cisplatin and gemcitabine, in a range of TNBC cell lines and particularly in olaparib-resistant models. In vivo assays on PDX validated the efficacy of dinaciclib which increased the sensitivity to olaparib of 5/6 models, including two olaparib-resistant and one BRCA1-WT model. However, no olaparib response improvement was observed in vivo with SR-4835. These data support that the implementation of CDK-inhibitors could be effective to sensitize TNBC to olaparib as well as possibly to cisplatin or gemcitabine.
Collapse
Affiliation(s)
- Esin Orhan
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Carolina Velazquez
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Imene Tabet
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Lise Fenou
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Geneviève Rodier
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Béatrice Orsetti
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - William Jacot
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France; Oncologie Clinique, Institut Du Cancer de Montpellier, Montpellier, France
| | - Claude Sardet
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France
| | - Charles Theillet
- Institut de Recherche en Cancérologie de Montpellier, IRCM, U1194, Montpellier University, INSERM, ICM, CNRS, Montpellier, France.
| |
Collapse
|
34
|
Molinuevo R, Menendez J, Cadle K, Ariqat N, Choy MK, Lagousis C, Thomas G, Strietzel C, Bubolz JW, Hinck L. Physiological DNA damage promotes functional endoreplication of mammary gland alveolar cells during lactation. Nat Commun 2024; 15:3288. [PMID: 38627401 PMCID: PMC11021458 DOI: 10.1038/s41467-024-47668-9] [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: 07/08/2022] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Lactation insufficiency affects many women worldwide. During lactation, a large portion of mammary gland alveolar cells become polyploid, but how these cells balance the hyperproliferation occurring during normal alveologenesis with terminal differentiation required for lactation is unknown. Here, we show that DNA damage accumulates due to replication stress during pregnancy, activating the DNA damage response. Modulation of DNA damage levels in vivo by intraductal injections of nucleosides or DNA damaging agents reveals that the degree of DNA damage accumulated during pregnancy governs endoreplication and milk production. We identify a mechanism involving early mitotic arrest through CDK1 inactivation, resulting in a heterogeneous alveolar population with regards to ploidy and nuclei number. The inactivation of CDK1 is mediated by the DNA damage response kinase WEE1 with homozygous loss of Wee1 resulting in decreased endoreplication, alveologenesis and milk production. Thus, we propose that the DNA damage response to replication stress couples proliferation and endoreplication during mammary gland alveologenesis. Our study sheds light on mechanisms governing lactogenesis and identifies non-hormonal means for increasing milk production.
Collapse
Affiliation(s)
- Rut Molinuevo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA, 95064, USA
| | - Julien Menendez
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA, 95064, USA
| | - Kora Cadle
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
| | - Nabeela Ariqat
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
| | - Marie Klaire Choy
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
| | - Cayla Lagousis
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
| | - Gwen Thomas
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA
| | | | - J W Bubolz
- Zoetis Inc., 333 Portage Street, Building 300, Kalamazoo, MI, 49007, USA
| | - Lindsay Hinck
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, 95064, USA.
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
35
|
Zhuang X, Liu B, Long J, Wang H, Yu J, Ji X, Li J, Zhu N, Li L, Chen Y, Liu Z, Zhao S. Machine-learning-based classification of diffuse large B-cell lymphoma patients by a 7-mRNA signature enriched with immune infiltration and cell cycle. Clin Transl Oncol 2024; 26:936-950. [PMID: 37783922 DOI: 10.1007/s12094-023-03326-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 09/12/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) exhibits remarkable heterogeneity but still remains undiagnosed in identifying the subpopulation of DLBCL to predict the prognosis and guide clinical treatment. METHODS Molecular subgroups were identified in gene expression data from GSE10846 by a consensus clustering algorithm. And gene set enrichment analysis, immune infiltration, and the proposed cell cycle algorithm were applied to explore the biological functions of different subtypes. Meanwhile, univariate and multivariate Cox regression analyses were used to evaluate independent prognostic factors of DLBCL. Finally, the prognostic model, including some key genes screened by Lasso regression, Random Forest algorithm, and point-biserial correlation, was constructed by an optimal classifier from seven machine learning algorithms and validated by another three external datasets (GSE34171, GSE87371, GSE31312). RESULTS Comprehensive genomic analysis of 1,143 DLBCL samples identify 2 molecularly, prognostically relevant subtypes: immune-enriched (IME) and cell-cycle-enriched (CCE). Then a new predictive model including seven key genes (SERPING1, TIMP2, NME1, DCTPP1, RFC4, POLE2, and SNRPD1) was developed with high prediction accuracy (88.6%) and strong predictive power (AUC = 0.973) based on the Support Vector Machine (SVM) algorithm in 414 patients from GSE10846. The predictive power was similar in another three testing sets (HR > 1.400, p < 0.05). CONCLUSION This model could evaluate survival independently with strong predictive power compared with other clinical risk factors. Our study constructed a reliable model to predict two new subtypes of DLBCL patients, which could guide the implementation of individualized treatment.
Collapse
Affiliation(s)
- Xujie Zhuang
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Bo Liu
- School of Mathematical and Computational Sciences, Massey University, Auckland, New Zealand
| | - Junqi Long
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Huina Wang
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Jiangyong Yu
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinchan Ji
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Jinmeng Li
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Nian Zhu
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Lujia Li
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Yuhaoran Chen
- School of Software Engineering, Faculty of Information Technology, Beijing University of Technology, Beijing, 100124, China
| | - Zhidong Liu
- Department of Thoracic Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Shuangtao Zhao
- Department of Breast Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute/Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
36
|
Bonatto MDS, Feltran GDS, Barbosa TP, Pereira DA, Santos SDS, Mendes PGJ, E Pessoa RS, Bezerra FJB, Zambuzzi WF, de Oliveira GJPL. Green tea and hyaluronic acid gel enhance fibroblast activation and improves the gingival healing post-third molar extraction. Sci Rep 2024; 14:7124. [PMID: 38531928 DOI: 10.1038/s41598-024-57821-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/21/2024] [Indexed: 03/28/2024] Open
Abstract
This study evaluates the effects of a green tea (Camellia sinensis) and hyaluronic acid gel on fibroblast activity and alveolar bone repair following third molar extractions. By examining the gene expression related to cell survival, proliferation, and angiogenesis, the study bridges in vitro findings with clinical outcomes in a split-mouth randomized trial. Human fibroblasts were exposed to the treatment gel, analysing gene expression through RT-qPCR. Twenty participants undergoing bilateral third molar extractions received the test gel on one side and a placebo on the other. Assessments included patient-reported outcomes, professional evaluations, and radiographic analyses at multiple postoperative intervals. The test gel significantly enhanced AKT, CDKs, and VEGF gene expressions, indicating a positive effect on angiogenesis and cell proliferation. Clinically, it resulted in reduced exudate, swelling, and secondary interventions, with radiographs showing improved alveolar bone density after 90 days. The green tea and hyaluronic acid gel significantly improves soft tissue and bone healing post-extraction, offering a promising adjunctive therapy for enhancing postoperative recovery. This gel represents a novel adjuvant treatment option for facilitating improved healing outcomes after third molar extractions, highlighting its potential utility in clinical dental practice.
Collapse
Affiliation(s)
- Mariana da Silva Bonatto
- School of Dentistry - Universidade Federal de Uberlândia (UFU), Pará, Av., 1760-1844, Uberlândia, MG, 38405-320, Brazil
| | - Geórgia da Silva Feltran
- UNESP - Campus de Botucatu - Instituto de Biociências, São Paulo State University, Rua Prof a. Dr a. Irina Delanova Gemtchujnicov, s/nº, Botucatu, SP, 18618-693, Brazil
| | - Thamires Prazeres Barbosa
- UNESP - Campus de Botucatu - Instituto de Biociências, São Paulo State University, Rua Prof a. Dr a. Irina Delanova Gemtchujnicov, s/nº, Botucatu, SP, 18618-693, Brazil
| | - Davisson Alves Pereira
- School of Dentistry - Universidade Federal de Uberlândia (UFU), Pará, Av., 1760-1844, Uberlândia, MG, 38405-320, Brazil
| | - Samara de Souza Santos
- School of Dentistry - Universidade Federal de Uberlândia (UFU), Pará, Av., 1760-1844, Uberlândia, MG, 38405-320, Brazil
| | - Pedro Gomes Junqueira Mendes
- School of Dentistry - Universidade Federal de Uberlândia (UFU), Pará, Av., 1760-1844, Uberlândia, MG, 38405-320, Brazil
| | | | - Fábio José Barbosa Bezerra
- UNESP - Campus de Botucatu - Instituto de Biociências, São Paulo State University, Rua Prof a. Dr a. Irina Delanova Gemtchujnicov, s/nº, Botucatu, SP, 18618-693, Brazil
| | - Willian Fernando Zambuzzi
- UNESP - Campus de Botucatu - Instituto de Biociências, São Paulo State University, Rua Prof a. Dr a. Irina Delanova Gemtchujnicov, s/nº, Botucatu, SP, 18618-693, Brazil.
| | | |
Collapse
|
37
|
Scheidecker B, Poulain S, Sugimoto M, Kido T, Kawanishi T, Miyajima A, Kim SH, Arakawa H, Kato Y, Nishikawa M, Danoy M, Sakai Y, Leclerc E. Dynamic, IPSC-derived hepatic tissue tri-culture system for the evaluation of liver physiology in vitro. Biofabrication 2024; 16:025037. [PMID: 38447229 DOI: 10.1088/1758-5090/ad30c5] [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: 10/12/2023] [Accepted: 03/06/2024] [Indexed: 03/08/2024]
Abstract
Availability of hepatic tissue for the investigation of metabolic processes is severely limited. While primary hepatocytes or animal models are widely used in pharmacological applications, a change in methodology towards more sustainable and ethical assays is highly desirable. Stem cell derived hepatic cells are generally regarded as a viable alternative for the above model systems, if current limitations in functionality and maturation can be overcome. By combining microfluidic organ-on-a-chip technology with individually differentiated, multicellular hepatic tissue fractions, we aim to improve overall functionality of hepatocyte-like cells, as well as evaluate cellular composition and interactions with non-parenchymal cell populations towards the formation of mature liver tissue. Utilizing a multi-omic approach, we show the improved maturation profiles of hepatocyte-like cells maintained in a dynamic microenvironment compared to standard tissue culture setups without continuous perfusion. In order to evaluate the resulting tissue, we employ single cell sequencing to distinguish formed subpopulations and spatial localization. While cellular input was strictly defined based on established differentiation protocols of parenchyma, endothelial and stellate cell fractions, resulting hepatic tissue was shown to comprise a complex mixture of epithelial and non-parenchymal fractions with specific local enrichment of phenotypes along the microchannel. Following this approach, we show the importance of passive, paracrine developmental processes in tissue formation. Using such complex tissue models is a crucial first step to develop stem cell-derivedin vitrosystems that can compare functionally with currently used pharmacological and toxicological applications.
Collapse
Affiliation(s)
- Benedikt Scheidecker
- CNRS UMI 2820, Institute of Industrial Science, University of Tokyo, 153-8505 Tokyo, Japan
| | - Stéphane Poulain
- Institute of Industrial Science, University of Tokyo, 153-8505 Tokyo, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, 997-0035 Yamagata, Japan
- Institute of Medical Science, Tokyo Medical University, 160-8402 Tokyo, Japan
| | - Taketomo Kido
- Institute for Quantitative Biosciences, University of Tokyo, 113-0032 Tokyo, Japan
| | - Takumi Kawanishi
- School of Pharmaceutical Sciences, Kanazawa University, 920-1102 Kanazawa, Japan
| | - Atsushi Miyajima
- Institute for Quantitative Biosciences, University of Tokyo, 113-0032 Tokyo, Japan
| | - Soo Hyeon Kim
- Institute of Industrial Science, University of Tokyo, 153-8505 Tokyo, Japan
| | - Hiroshi Arakawa
- School of Pharmaceutical Sciences, Kanazawa University, 920-1102 Kanazawa, Japan
| | - Yukio Kato
- School of Pharmaceutical Sciences, Kanazawa University, 920-1102 Kanazawa, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, University of Tokyo, 113-8654 Tokyo, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, University of Tokyo, 113-8654 Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, University of Tokyo, 113-8654 Tokyo, Japan
| | - Eric Leclerc
- CNRS UMI 2820, Institute of Industrial Science, University of Tokyo, 153-8505 Tokyo, Japan
- CNRS UMR 7338, Laboratoire de Biomécanique et Bioingénierie, Université de Technologies de Compiègne, 60203 Compiègne, France
| |
Collapse
|
38
|
Huang YT, Hesting LL, Calvi BR. An unscheduled switch to endocycles induces a reversible senescent arrest that impairs growth of the Drosophila wing disc. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.14.585098. [PMID: 38559130 PMCID: PMC10980049 DOI: 10.1101/2024.03.14.585098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
A programmed developmental switch to G / S endocycles results in tissue growth through an increase in cell size. Unscheduled, induced endocycling cells (iECs) promote wound healing but also contribute to cancer. Much remains unknown, however, about how these iECs affect tissue growth. Using the D. melanogasterwing disc as model, we find that populations of iECs initially increase in size but then subsequently undergo a heterogenous arrest that causes severe tissue undergrowth. iECs acquired DNA damage and activated a Jun N-terminal kinase (JNK) pathway, but, unlike other stressed cells, were apoptosis-resistant and not eliminated from the epithelium. Instead, iECs entered a JNK-dependent and reversible senescent-like arrest. Senescent iECs promoted division of diploid neighbors, but this compensatory proliferation did not rescue tissue growth. Our study has uncovered unique attributes of iECs and their effects on tissue growth that have important implications for understanding their roles in wound healing and cancer.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Lauren L. Hesting
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| | - Brian R. Calvi
- Department of Biology, Simon Cancer Center, Indiana University, Bloomington, IN 47405
| |
Collapse
|
39
|
Zhao Y, Yang Y, Wu X, Zhang L, Cai X, Ji J, Chen S, Vera A, Boström KI, Yao Y. CDK1 inhibition reduces osteogenesis in endothelial cells in vascular calcification. JCI Insight 2024; 9:e176065. [PMID: 38456502 PMCID: PMC10972591 DOI: 10.1172/jci.insight.176065] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/19/2024] [Indexed: 03/09/2024] Open
Abstract
Vascular calcification is a severe complication of cardiovascular diseases. Previous studies demonstrated that endothelial lineage cells transitioned into osteoblast-like cells and contributed to vascular calcification. Here, we found that inhibition of cyclin-dependent kinase (CDK) prevented endothelial lineage cells from transitioning to osteoblast-like cells and reduced vascular calcification. We identified a robust induction of CDK1 in endothelial cells (ECs) in calcified arteries and showed that EC-specific gene deletion of CDK1 decreased the calcification. We found that limiting CDK1 induced E-twenty-six specific sequence variant 2 (ETV2), which was responsible for blocking endothelial lineage cells from undergoing osteoblast differentiation. We also found that inhibition of CDK1 reduced vascular calcification in a diabetic mouse model. Together, the results highlight the importance of CDK1 suppression and suggest CDK1 inhibition as a potential option for treating vascular calcification.
Collapse
Affiliation(s)
- Yan Zhao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Yang Yang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xiuju Wu
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Li Zhang
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Xinjiang Cai
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Jaden Ji
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Sydney Chen
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Abigail Vera
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Kristina I. Boström
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- The Molecular Biology Institute at UCLA, Los Angeles, California, USA
| | - Yucheng Yao
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
40
|
Kang X, Chen H, Zhou Z, Tu S, Cui B, Li Y, Dong S, Zhang Q, Xu Y. Targeting Cyclin-Dependent Kinase 1 Induces Apoptosis and Cell Cycle Arrest of Activated Hepatic Stellate Cells. Adv Biol (Weinh) 2024; 8:e2300403. [PMID: 38103005 DOI: 10.1002/adbi.202300403] [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: 08/04/2023] [Revised: 11/08/2023] [Indexed: 12/17/2023]
Abstract
Liver fibrosis is the integral process of chronic liver diseases caused by multiple etiologies and characterized by excessive deposition of extracellular matrix (ECM). During liver fibrosis, hepatic stellate cells (HSCs) transform into a highly proliferative, activated state, producing various cytokines, chemokines, and ECM. However, the precise mechanisms that license HSCs into the highly proliferative state remain unclear. Cyclin-dependent kinase 1 (CDK1) is a requisite event for the transition of the G1/S and G2/M phases in eukaryotic cells. In this study, it is demonstrated that CDK1 and its activating partners, Cyclin A2 and Cyclin B1, are upregulated in both liver fibrosis/cirrhosis patient specimens and the murine hepatic fibrosis models, especially in activated HSCs. In vitro, CDK1 is upregulated in spontaneously activated HSCs, and inhibiting CDK1 with specific small-molecule inhibitors (CGP74514A, RO-3306, or Purvalanol A) orshort hairpin RNAs (shRNAs) resulted in HSC apoptosis and cell cycle arrest by regulating Survivin expression. Above all, it is illustrated that increased CDK1 expression licenses the HSCs into a highly proliferative state and can serve as a potential therapeutic target in liver fibrosis.
Collapse
Affiliation(s)
- Xinmei Kang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Huaxin Chen
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Zhuowei Zhou
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Silin Tu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Bo Cui
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Yanli Li
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Shuai Dong
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Qi Zhang
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
- Cell-gene Therapy Translational Medicine Research Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| | - Yan Xu
- Biotherapy Centre, the Third Affiliated Hospital, Sun Yat-sen University, 600# Tianhe Road, Guangzhou, 510630, China
| |
Collapse
|
41
|
Dasgupta D, Mahadev Bhat S, Creighton C, Cortes C, Delmotte P, Sieck GC. Molecular mechanisms underlying TNFα-induced mitochondrial fragmentation in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2024; 326:L190-L205. [PMID: 38084427 PMCID: PMC11280718 DOI: 10.1152/ajplung.00198.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Abstract
Tumor necrosis factor α (TNFα), a proinflammatory cytokine, plays a significant role in mediating the effects of acute inflammation in response to allergens, pollutants, and respiratory infections. Previously, we showed that acute exposure to TNFα induces mitochondrial fragmentation in human airway smooth muscle (hASM) cells, which is associated with increased expression of dynamin-related protein 1 (DRP1). Phosphorylation of DRP1 at serine 616 (pDRP1S616) promotes its translocation and binding to the outer mitochondrial membrane (OMM) and mediates mitochondrial fragmentation. Previously, we reported that TNFα exposure triggers protein unfolding and triggers an endoplasmic reticulum (ER) stress response involving phosphorylation of inositol-requiring enzyme 1α (pIRE1α) at serine 724 (pIRE1αS724) and subsequent splicing of X-box binding protein 1 (XBP1s) in hASM cells. We hypothesize that TNFα-mediated activation of the pIRE1αS724/XBP1s ER stress pathway in hASM cells transcriptionally activates genes that encode kinases responsible for pDRP1S616 phosphorylation. Using 3-D confocal imaging of MitoTracker green-labeled mitochondria, we found that TNFα treatment for 6 h induces mitochondrial fragmentation in hASM cells. We also confirmed that 6 h TNFα treatment activates the pIRE1α/XBP1s ER stress pathway. Using in silico analysis and ChIP assay, we showed that CDK1 and CDK5, kinases involved in the phosphorylation of pDRP1S616, are transcriptionally targeted by XBP1s. TNFα treatment increased the binding affinity of XBP1s on the promoter regions of CDK1 and CDK5, and this was associated with an increase in pDRP1S616 and mitochondria fragmentation. This study reveals a new underlying molecular mechanism for TNFα-induced mitochondrial fragmentation in hASM cells.NEW & NOTEWORTHY Airway inflammation is increasing worldwide. Proinflammatory cytokines mediate an adaptive mechanism to overcome inflammation-induced cellular stress. Previously, we reported that TNFα mediates hASM cellular responses, leading to increased force and ATP consumption associated with increased O2 consumption, and oxidative stress. This study indicates that TNFα induces ER stress, which induces mitochondrial fragmentation via pIRE1αS724/XBP1s mediated CDK1/5 upregulation and pDRP1S616 phosphorylation. Mitochondrial fragmentation may promote hASM mitochondrial biogenesis to maintain healthy mitochondrial pool.
Collapse
Affiliation(s)
- Debanjali Dasgupta
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Sanjana Mahadev Bhat
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Claire Creighton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Catherin Cortes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Philippe Delmotte
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
42
|
Lv X, An Y. Bioinformatics-based Identification of Ferroptosis-related Genes and their Diagnostic Value in Gestational Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2024; 24:1611-1621. [PMID: 38347799 DOI: 10.2174/0118715303275367240103102801] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/14/2023] [Accepted: 12/07/2023] [Indexed: 10/22/2024]
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) is considered a risk factor for heart metabolic disorder in future mothers and offspring. Ferroptosis is a new type of programmed cell death, which may participate in the occurrence and development of GDM. OBJECTIVE This study aims to identify ferroptosis-related genes in GDM by bioinformatics methods and to explore their clinical diagnostic value. METHODS The dataset GSE103552 was analyzed using the Gene Expression Omnibus (GEO) database to screen for differentially expressed genes (DEGs) in GDM. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and proteinprotein interaction (PPI) network were performed. Gene sets for ferroptosis were retrieved in MSigDB and GSVA gene set analysis was performed on the database. Finally, logistic regression was performed to differentiate between GDM patients and controls to screen for diagnostic markers. RESULTS A total of 179 DEGs were identified in the expression profile of GDM. GO and KEGG enrichment analysis revealed significant enrichment in the TGF-β, p53 signaling pathway, platelet activation, glutathione metabolism, sensory perception of taste, and leukocyte and vascular endothelial cell migration regulation. DEGs (n = 107) associated with the ferroptosis gene set were screened by GSVA analysis. The screened DEGs for disease and DEGs for ferroptosis scores were intersected and 35 intersected genes were identified. PPI identified two key genes associated with GDM as CCNB2 and CDK1. Wilcox-test showed low expression of CCNB2 and CDK1 in GDM. The area under the ROC curve (AUC) of the CCNB2 and CDK1 prognostic model was 0.822. CONCLUSION The genes associated with ferroptosis in GDM were CCNB2 and CDK1, which can be used as valid indicators for the diagnosis of GDM.
Collapse
Affiliation(s)
- Xiaomei Lv
- Department of Obstetrics, Jinan, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yujun An
- Department of Obstetrics, Jinan, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
43
|
Jacob TV, Doshi GM. A Mini-review on Helicobacter pylori with Gastric Cancer and Available Treatments. Endocr Metab Immune Disord Drug Targets 2024; 24:277-290. [PMID: 37622707 DOI: 10.2174/1871530323666230824161901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/26/2023]
Abstract
Helicobacter pylori (H. pylori) is the most thoroughly researched etiological component for stomach inflammation and malignancies. Even though there are conventional recommendations and treatment regimens for eradicating H. pylori, failure rates continue to climb. Antibiotic resistance contributes significantly to misdiagnoses, false positive results, and clinical failures, all of which raise the chance of infection recurrence. This review aims to explore the molecular mechanisms underlying drug resistance in H. pylori and discuss novel approaches for detecting genotypic resistance. Modulation of drug uptake/ efflux, biofilm, and coccoid development. Newer genome sequencing approaches capable of detecting H. pylori genotypic resistance are presented. Prolonged infection in the stomach causes major problems such as gastric cancer. The review discusses how H. pylori causes stomach cancer, recent biomarkers such as miRNAs, molecular pathways in the development of gastric cancer, and diagnostic methods and clinical trials for the disease. Efforts have been made to summarize the recent advancements made toward early diagnosis and novel therapeutic approaches for H. pylori-induced gastric cancer.
Collapse
Affiliation(s)
- Teresa V Jacob
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
44
|
Gall L, Duckworth C, Jardi F, Lammens L, Parker A, Bianco A, Kimko H, Pritchard DM, Pin C. Homeostasis, injury, and recovery dynamics at multiple scales in a self-organizing mouse intestinal crypt. eLife 2023; 12:e85478. [PMID: 38063302 PMCID: PMC10789491 DOI: 10.7554/elife.85478] [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: 12/09/2022] [Accepted: 12/07/2023] [Indexed: 01/16/2024] Open
Abstract
The maintenance of the functional integrity of the intestinal epithelium requires a tight coordination between cell production, migration, and shedding along the crypt-villus axis. Dysregulation of these processes may result in loss of the intestinal barrier and disease. With the aim of generating a more complete and integrated understanding of how the epithelium maintains homeostasis and recovers after injury, we have built a multi-scale agent-based model (ABM) of the mouse intestinal epithelium. We demonstrate that stable, self-organizing behaviour in the crypt emerges from the dynamic interaction of multiple signalling pathways, such as Wnt, Notch, BMP, ZNRF3/RNF43, and YAP-Hippo pathways, which regulate proliferation and differentiation, respond to environmental mechanical cues, form feedback mechanisms, and modulate the dynamics of the cell cycle protein network. The model recapitulates the crypt phenotype reported after persistent stem cell ablation and after the inhibition of the CDK1 cycle protein. Moreover, we simulated 5-fluorouracil (5-FU)-induced toxicity at multiple scales starting from DNA and RNA damage, which disrupts the cell cycle, cell signalling, proliferation, differentiation, and migration and leads to loss of barrier integrity. During recovery, our in silico crypt regenerates its structure in a self-organizing, dynamic fashion driven by dedifferentiation and enhanced by negative feedback loops. Thus, the model enables the simulation of xenobiotic-, in particular chemotherapy-, induced mechanisms of intestinal toxicity and epithelial recovery. Overall, we present a systems model able to simulate the disruption of molecular events and its impact across multiple levels of epithelial organization and demonstrate its application to epithelial research and drug development.
Collapse
Affiliation(s)
- Louis Gall
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - Carrie Duckworth
- Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Ferran Jardi
- Preclinical Sciences and Translational Safety, JanssenBeerseBelgium
| | - Lieve Lammens
- Preclinical Sciences and Translational Safety, JanssenBeerseBelgium
| | - Aimee Parker
- Gut Microbes and Health Programme, Quadram InstituteNorwichUnited Kingdom
| | - Ambra Bianco
- Clinical Pharmacology and Safety Sciences, AstraZenecaCambridgeUnited Kingdom
| | - Holly Kimko
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - David Mark Pritchard
- Institute of Systems, Molecular and Integrative Biology, University of LiverpoolLiverpoolUnited Kingdom
| | - Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| |
Collapse
|
45
|
Tanaka T, Miyakoshi Y, Kobayashi Y, Xiaolong S, Daiyang Y, Ochi H, Sato S, Kato T, Yoshii T, Okawa A, Kaldis P, Inose H. Regulation of Osteoblast to Osteocyte Differentiation by Cyclin-Dependent Kinase-1. Adv Biol (Weinh) 2023; 7:e2300136. [PMID: 37424388 DOI: 10.1002/adbi.202300136] [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: 04/07/2023] [Revised: 06/16/2023] [Indexed: 07/11/2023]
Abstract
Osteocytes have recently been identified as a new regulator of bone remodeling, but the detailed mechanism of their differentiation from osteoblasts remains unclear. The purpose of this study is to identify cell cycle regulators involved in the differentiation of osteoblasts into osteocytes and determine their physiological significance. The study uses IDG-SW3 cells as a model for the differentiation from osteoblasts to osteocytes. Among the major cyclin-dependent kinases (Cdks), Cdk1 is most abundantly expressed in IDG-SW3 cells, and its expression is down-regulated during differentiation into osteocytes. Inhibition of CDK1 activity reduces IDG-SW3 cell proliferation and differentiation into osteocytes. Osteocyte and Osteoblast-specific Cdk1 knockout in mice (Dmp1-Cdk1KO ) results in trabecular bone loss. Pthlh expression increases during differentiation, but inhibiting CDK1 activity reduces Pthlh expression. Parathyroid hormone-related protein concentration is reduced in the bone marrow of Dmp1-Cdk1KO mice. Four weeks of Parathyroid hormone administration partially recovers the trabecular bone loss in Dmp1-Cdk1KO mice. These results demonstrate that Cdk1 plays an essential role in the differentiation from osteoblast to osteocyte and the acquisition and maintenance of bone mass. The findings contribute to a better understanding of the mechanisms of bone mass regulation and can help develop efficient therapeutic strategies for osteoporosis treatment.
Collapse
Affiliation(s)
- Tomoyuki Tanaka
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
- Department of Orthopaedic Surgery, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minamikoshigaya, Koshigaya-shi, Saitama, 343-8555, Japan
| | - Yuri Miyakoshi
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yutaka Kobayashi
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Sun Xiaolong
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Yu Daiyang
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Hiroki Ochi
- Department of Rehabilitation for Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, 4-1 Namiki, Tokorozawa, Saitama, 359-8555, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Tsuyoshi Kato
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Toshitaka Yoshii
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Atsushi Okawa
- Department of Orthopaedics, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC) Box 50332, Malmö, SE-202 13, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, Malmö, SE-202 13, Sweden
| | - Hiroyuki Inose
- Department of Orthopaedic Surgery, Dokkyo Medical University Saitama Medical Center, 2-1-50 Minamikoshigaya, Koshigaya-shi, Saitama, 343-8555, Japan
- Department of Orthopedic and Trauma Research, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan
| |
Collapse
|
46
|
Zhang W, Li K, Li S, Lv R, Ma J, Yin P, Li L, Sun N, Chen Y, Lu L, Li Y, Zhang Q, Yan H. High-throughput sequencing reveals hub genes for human early embryonic development arrest in vitro fertilization: a pilot study. Front Physiol 2023; 14:1279559. [PMID: 38033342 PMCID: PMC10684309 DOI: 10.3389/fphys.2023.1279559] [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: 08/18/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
Many clinical studies have shown that embryos of in vitro fertilization (IVF) are often prone to developmental arrest, which leads to recurrent failure of IVF treatment. Early embryonic arrest has always been an urgent clinical problem in assisted reproduction centers. However, the molecular mechanisms underlying early embryonic development arrest remain largely unknown. The objective of this study is to investigate potential candidate hub genes and key signaling pathways involved in early stages of embryonic development. RNA-seq analysis was performed on normal and arrest embryos to study the changes of gene expression during early embryonic development. A total of 520 genes exhibiting differential expression were identified, with 174 genes being upregulated and 346 genes being downregulated. Upregulated genes show enrichment in biosynthesis, cellular proliferation and differentiation, and epigenetic regulation. While downregulated genes exhibit enrichment in transcriptional activity, epigenetic regulation, cell cycle progression, cellular proliferation and ubiquitination. The STRING (search tool for the retravel of interacting genes/proteins) database was utilized to analyze protein-protein interactions among these genes, aiming to enhance comprehension of the potential role of these differentially expressed genes (DEGs). A total of 22 hub genes (highly connected genes) were identified among the DEGs using Cytoscape software. Of these, ERBB2 and VEGFA were upregulated, while the remaining 20 genes (CCNB1, CCNA2, DICER1, NOTCH1, UBE2B, UBE2N, PRMT5, UBE2D1, MAPK3, SOX9, UBE2C, UB2D2, EGF, ACTB, UBA52, SHH, KRAS, UBE2E1, ADAM17 and BRCA2) were downregulated. These hub genes are associated with crucial biological processes such as ubiquitination, cellular senescence, cell proliferation and differentiation, and cell cycle. Among these hub genes, CCNA2 and CCNB1 may be involved in controlling cell cycle, which are critical process in early embryonic development.
Collapse
Affiliation(s)
- Wuwen Zhang
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shifeng Li
- Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Rong Lv
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Ma
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Yin
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ningyu Sun
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Chen
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinhua Zhang
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Abstract
The liver's unique chromosomal variations, including polyploidy and aneuploidy, influence hepatocyte identity and function. Among the most well-studied mammalian polyploid cells, hepatocytes exhibit a dynamic interplay between diploid and polyploid states. The ploidy state is dynamic as hepatocytes move through the "ploidy conveyor," undergoing ploidy reversal and re-polyploidization during proliferation. Both diploid and polyploid hepatocytes actively contribute to proliferation, with diploids demonstrating an enhanced proliferative capacity. This enhanced potential positions diploid hepatocytes as primary drivers of liver proliferation in multiple contexts, including homeostasis, regeneration and repopulation, compensatory proliferation following injury, and oncogenic proliferation. This review discusses the influence of ploidy variations on cellular activity. It presents a model for ploidy-associated hepatocyte proliferation, offering a deeper understanding of liver health and disease with the potential to uncover novel treatment approaches.
Collapse
Affiliation(s)
- Sierra R. Wilson
- Department of Pathology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew W. Duncan
- Department of Pathology, McGowan Institute for Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Erguven M, Kilic S, Karaca E, Diril MK. Genetic complementation screening and molecular docking give new insight on phosphorylation-dependent Mastl kinase activation. J Biomol Struct Dyn 2023; 41:8241-8253. [PMID: 36270968 DOI: 10.1080/07391102.2022.2131627] [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: 08/26/2022] [Accepted: 09/26/2022] [Indexed: 10/24/2022]
Abstract
Mastl is a mitotic kinase that is essential for error-free chromosome segregation. It is an atypical member of AGC kinase family, possessing a unique non-conserved middle region. The mechanism of Mastl activation has been studied extensively in vitro. Phosphorylation of several residues were identified to be crucial for activation. These sites correspond to T193 and T206 in the activation loop and S861 in the C-terminal tail of mouse Mastl. To date, the significance of these phosphosites was not confirmed in intact mammalian cells. Here, we utilize a genetic complementation approach to determine the essentials of mammalian Mastl kinase activation. We used tamoxifen-inducible conditional knockout mouse embryonic fibroblasts to delete endogenous Mastl and screened various mutants for their ability to complement its loss. S861A mutant was able to complement endogenous Mastl loss. In parallel, we performed computational molecular docking studies to evaluate the significance of this residue for kinase activation. Our in-depth sequence and structure analysis revealed that Mastl pS861 does not belong to a conformational state, where the phosphoresidue contributes to C-tail docking. C-tail of Mastl is relatively short and it lacks a hydrophobic (HF) motif that would otherwise help its anchoring over N-lobe, required for the final steps of kinase activation. Our results show that phosphorylation of Mastl C-tail turn motif (S861) is dispensable for kinase function in cellulo.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mehmet Erguven
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Seval Kilic
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ezgi Karaca
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - M Kasim Diril
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
- Department of Medical Biology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
49
|
Hatton IA, Galbraith ED, Merleau NSC, Miettinen TP, Smith BM, Shander JA. The human cell count and size distribution. Proc Natl Acad Sci U S A 2023; 120:e2303077120. [PMID: 37722043 PMCID: PMC10523466 DOI: 10.1073/pnas.2303077120] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/24/2023] [Indexed: 09/20/2023] Open
Abstract
Cell size and cell count are adaptively regulated and intimately linked to growth and function. Yet, despite their widespread relevance, the relation between cell size and count has never been formally examined over the whole human body. Here, we compile a comprehensive dataset of cell size and count over all major cell types, with data drawn from >1,500 published sources. We consider the body of a representative male (70 kg), which allows further estimates of a female (60 kg) and 10-y-old child (32 kg). We build a hierarchical interface for the cellular organization of the body, giving easy access to data, methods, and sources (https://humancelltreemap.mis.mpg.de/). In total, we estimate total body counts of ≈36 trillion cells in the male, ≈28 trillion in the female, and ≈17 trillion in the child. These data reveal a surprising inverse relation between cell size and count, implying a trade-off between these variables, such that all cells within a given logarithmic size class contribute an equal fraction to the body's total cellular biomass. We also find that the coefficient of variation is approximately independent of mean cell size, implying the existence of cell-size regulation across cell types. Our data serve to establish a holistic quantitative framework for the cells of the human body, and highlight large-scale patterns in cell biology.
Collapse
Affiliation(s)
- Ian A. Hatton
- Max Planck Institute for Mathematics in the Sciences, Leipzig04103, Germany
- Department of Earth and Planetary Sciences, McGill University, Montreal, QuebecH3A 0E8, Canada
| | - Eric D. Galbraith
- Department of Earth and Planetary Sciences, McGill University, Montreal, QuebecH3A 0E8, Canada
- ICREA, Barcelona08010, Spain
| | - Nono S. C. Merleau
- Max Planck Institute for Mathematics in the Sciences, Leipzig04103, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, University of Leipzig, D-04105Leipzig, Germany
| | - Teemu P. Miettinen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Benjamin McDonald Smith
- Department of Medicine, McGill University Health Centre Research Institute, Montreal, QuebecH4A 3S5, Canada
- Department of Medicine, Columbia University Medical Center, New York, NY10032
| | | |
Collapse
|
50
|
Pluta AJ, Studniarek C, Murphy S, Norbury CJ. Cyclin-dependent kinases: Masters of the eukaryotic universe. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 15:e1816. [PMID: 37718413 PMCID: PMC10909489 DOI: 10.1002/wrna.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/19/2023]
Abstract
A family of structurally related cyclin-dependent protein kinases (CDKs) drives many aspects of eukaryotic cell function. Much of the literature in this area has considered individual members of this family to act primarily either as regulators of the cell cycle, the context in which CDKs were first discovered, or as regulators of transcription. Until recently, CDK7 was the only clear example of a CDK that functions in both processes. However, new data points to several "cell-cycle" CDKs having important roles in transcription and some "transcriptional" CDKs having cell cycle-related targets. For example, novel functions in transcription have been demonstrated for the archetypal cell cycle regulator CDK1. The increasing evidence of the overlap between these two CDK types suggests that they might play a critical role in coordinating the two processes. Here we review the canonical functions of cell-cycle and transcriptional CDKs, and provide an update on how these kinases collaborate to perform important cellular functions. We also provide a brief overview of how dysregulation of CDKs contributes to carcinogenesis, and possible treatment avenues. This article is categorized under: RNA Interactions with Proteins and Other Molecules > RNA-Protein Complexes RNA Processing > 3' End Processing RNA Processing > Splicing Regulation/Alternative Splicing.
Collapse
Affiliation(s)
| | | | - Shona Murphy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Chris J. Norbury
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| |
Collapse
|