201
|
Satbhai KM, Marques ES, Ranjan R, Timme-Laragy AR. Single-cell RNA sequencing reveals tissue-specific transcriptomic changes induced by perfluorooctanesulfonic acid (PFOS) in larval zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137515. [PMID: 39947082 PMCID: PMC12038816 DOI: 10.1016/j.jhazmat.2025.137515] [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: 08/22/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 04/16/2025]
Abstract
Perfluorooctanesulfonic acid (PFOS) elicits adverse effects on numerous organs and developmental processes but the mechanisms underlying these effects are not well understood. Here, we use single-cell RNA-sequencing to assess tissue-specific transcriptomic changes in zebrafish (Danio rerio) larvae exposed to 16 µM PFOS or dimethylsulfoxide (0.01 %) from 3-72 h post fertilization (hpf). Data analysis was multi-pronged and included pseudo-bulk, untargeted clustering, informed pathway queries, and a cluster curated for hepatocyte biomarkers (fabp10a, and apoa2). Overall, 8.63 % (2390/27698) genes were significantly differentially expressed. Results from untargeted analysis revealed 22 distinct clusters that were manually annotated to specific tissues using a weight-of-evidence approach. The clusters with the highest number of significant differentially expressed genes (DEGs) were digestive organs, muscle, and otolith. Additionally, we assessed the distribution of pathway-specific genes known to be involved in PFOS toxicity: the PPAR pathway, β-oxidation of fatty acids, the Nfe2l2 pathway, and epigenetic modifications by DNA methylation, across clusters and identified the blood-related tissue to be the most sensitive. The curated hepatocyte cluster showed 220 significant DEGs and was enriched for the Notch signaling pathway. These findings provide insights into both established and novel sensitive target tissues and molecular mechanisms of developmental toxicity of PFOS.
Collapse
Affiliation(s)
- Kruuttika M Satbhai
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Emily S Marques
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Ravi Ranjan
- Genomics Resource Laboratory, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
202
|
Li G, Chen W, Liu D, Tang S. Recent advances in medicinal chemistry strategies for the development of METTL3 inhibitors. Eur J Med Chem 2025; 290:117560. [PMID: 40147343 DOI: 10.1016/j.ejmech.2025.117560] [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: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
N6-methyladenosine (m6A), the most abundant RNA modification in eukaryotic cells, exerts a critical influence on RNA function and gene expression. It has attracted considerable attention within the rapidly evolving field of epitranscriptomics. METTL3 is a key enzyme for m6A modification and is essential for maintaining normal m6A levels. High expression of METTL3 is closely associated with various cancers, including gastric cancer, liver cancer, and leukemia. Inhibiting METTL3 has shown potential in slowing cancer progression, thereby driving the development of METTL3 inhibitors. In this work, we summarize recent advancements in the development of METTL3 inhibitor, with a focus on medicinal chemistry strategies employed during discovery and optimization phases. We explore the application of structure-activity relationship (SAR) studies and protein-targeted degradation techniques, while addressing key challenges associated with their characterization and clinical translation. This review underscores the therapeutic potential of METTL3 inhibitors in modulating epitranscriptomic pathways and aims to offer perspectives for future research in this rapidly evolving field.
Collapse
Affiliation(s)
- Gengwu Li
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Chen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shibing Tang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; State Key Laboratory of Respiratory Disease, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
203
|
Zhang Q, Li L, Li S, Zhou X. Small molecule compounds targeting G9a/GLP: Recent advances and perspectives. Eur J Med Chem 2025; 290:117525. [PMID: 40121866 DOI: 10.1016/j.ejmech.2025.117525] [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: 01/08/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
As an important member of the histone methyltransferase family, G9a/GLP has been shown to be closely related to the occurrence and development of various diseases, such as tumors, fibrosis, and malaria. Selective small molecule inhibitors of G9a/GLP were first reported in 2007, and over the decade since then, more than 40 different types of G9a modulators have been developed. Classification by binding site includes s-adenosylmethionine (SAM)-competitive inhibitors and substrate-competitive inhibitors. According to the mechanism of action, these compounds can be divided into reversible inhibitors, irreversible inhibitors, dual inhibitors, degraders, etc. In this paper, we systematically reviewed the discovery methods, design strategies, structural optimization processes, binding modes, biological activity data, and pharmacokinetic properties of small molecules targeting G9a/GLP. This paper analyzed the challenges and opportunities in the development of small molecule compounds targeting G9a/GLP, aiming to offer valuable insights and perspectives for pharmaceutical researchers.
Collapse
Affiliation(s)
- Qiangsheng Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Lu Li
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Siyan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 17#3rd Section, Ren Min South Road, Chengdu, 610041, China
| | - Xianli Zhou
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
204
|
Chen W, Zhuang X, Chen Y, Shen L, Yang H, Wang M, Pan G, Tan J, Pan X, Feng S, Yuan K, Zhang XY, Yang P. Discovery of potent and selective CDK2 inhibitors with high safety and favorable bioavailability for the treatment of cancer. Eur J Med Chem 2025; 290:117503. [PMID: 40107208 DOI: 10.1016/j.ejmech.2025.117503] [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: 01/17/2025] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Targeting cyclin-dependent kinases (CDKs) to inhibit the cell proliferation is considered as a promising strategy for the treatment of cancer, and the success of selective CDK4/6 inhibitors proves this concept. CDK2 plays an important role in the cell cycle and proliferation for the CCNE1-amplifed cancers and CDK4/6 inhibitors resistant breast cancers. Therefore, selective inhibition of CDK2 become research hotspots. In our work, we achieved a potent and selective CDK2 inhibitor 46 through virtual screening and systematic structural modification. Compound 46 could arrest cell cycle, promote apoptosis, and induce senescence-related phenotypes for CCNE1-amplifed ovarian cancer OVCAR3 cell line, and also displayed potent antitumor activity against OVCAR3 xenografts. Furthermore, 46 hold promise in overcoming resistance to CDK4/6 inhibitor. More significantly, 46 exhibited great safety properties and favorable pharmacokinetic profiles in vivo. All these results demonstrated that 46 was a potential candidate of novel anticancer drugs.
Collapse
Affiliation(s)
- Weijiao Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xujie Zhuang
- State Key Laboratory of Natural Medicines, 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, 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, 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, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Minjie Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Guoyong Pan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jinke Tan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xu Pan
- State Key Laboratory of Natural Medicines, 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, 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, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiao-Yu Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
205
|
Song Y, Li J, Luan X, Li A, Liu N, Wu ZH, Yang W, Gao W, Zheng X, Zhang XQ. OsCHR728 encodes a chromatin remodeling factor involved in seed size and grain chalkiness in rice. Gene 2025; 951:149396. [PMID: 40058712 DOI: 10.1016/j.gene.2025.149396] [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: 10/24/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
The Imitation Switch (ISWI)ATP-dependent chromatin remodeling factor proteins regulate various developmental processes, spanning from flowering to stress response. However, researches on the roles of ISWI subfamily genes in rice have been limited. This study investigated the expression profile of the rice chromatin remodeler OsCHR728, encoding an ISWI protein, across various tissues and developing stages. Our findings reveal that OsCHR728 is highly accumulated during early stage of the panicle development. We generated OsCHR728 knockout (KO) lines in rice using CRISPR/Cas9 technology. These mutant lines displayed smaller grain size compared to the wild type (Zhonghua 11, ZH11). Expression analysis revealed a significant downregulation of the transcript levels of five genes associated with cell cycle regulation in KO grains compared to the wild type, consistent with the reduced cell number in the mutant grains. Additionally, total free amino acid levels were higher in the KO mutant compared to the wild type, consequently enhancing the nutritional quality of the KO mutant grains. The mature endosperm of the KO mutant exhibited a reduced percentage of chalky grains and less chalkiness, suggesting an improvement in the appearance quality of the KO mutant. These results suggest that chromatin remodeling factor OsCHR728 plays a role in grain development, potentially providing a new avenue to enhance both the appearance and nutritional quality of rice cultivars.
Collapse
Affiliation(s)
- Yuxin Song
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China
| | - Jieni Li
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China; Rice Research Institute, Guangxi Academy of Agricultural Sciences/Guangxi Key Laboratory of Rice Genetics and Breeding, Nanning, China
| | - Xin Luan
- Rice Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Ao Li
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China
| | - Na Liu
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China
| | - Zhi-Hao Wu
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Weifeng Yang
- College of Agriculture, South China Agricultural University, Guangzhou, China
| | - Wanzhen Gao
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China
| | - Xia Zheng
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, China.
| | - Xiang-Qian Zhang
- Department of Horticulture, College of Agricultural and Biological Engineering, Foshan University, Foshan, China.
| |
Collapse
|
206
|
Zang JL, Gibson D, Zheng AM, Shi W, Gillies JP, Stein C, Drerup CM, DeSantis ME. CCSer2 gates dynein activity at the cell periphery. J Cell Biol 2025; 224:e202406153. [PMID: 40261303 PMCID: PMC12013514 DOI: 10.1083/jcb.202406153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/07/2025] [Accepted: 03/13/2025] [Indexed: 04/24/2025] Open
Abstract
Cytoplasmic dynein-1 (dynein) is a microtubule-associated, minus end-directed motor that traffics hundreds of different cargos. Dynein must discriminate between cargos and traffic them at the appropriate time from the correct cellular region. How dynein's trafficking activity is regulated in time or cellular space remains poorly understood. Here, we identify CCSer2 as the first known protein to gate dynein activity in the spatial dimension. CCSer2 promotes the migration of developing zebrafish primordium cells, macrophages, and cultured human cells by facilitating the trafficking of cargos that are acted on by peripherally localized dynein. Our data suggest that CCSer2 disfavors the interaction between dynein and its regulator Ndel1 at the cell edge, resulting in localized dynein activation. These findings support a model where the spatial specificity of dynein is achieved by the localization of proteins that trigger Ndel1's release from dynein. We propose that CCSer2 defines a broader class of proteins that activate dynein in distinct microenvironments via regulating Ndel1-dynein interaction.
Collapse
Affiliation(s)
- Juliana L. Zang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daytan Gibson
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ann-Marie Zheng
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Wanjing Shi
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - John P. Gillies
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Chris Stein
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Catherine M. Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Morgan E. DeSantis
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
207
|
Shelford J, Burgess SG, Rostkova E, Richards MW, Larocque G, Sampson J, Tiede C, Fielding AJ, Daviter T, Tomlinson DC, Calabrese AN, Pfuhl M, Bayliss R, Royle SJ. Structural characterization and inhibition of the interaction between ch-TOG and TACC3. J Cell Biol 2025; 224:e202407002. [PMID: 40105698 PMCID: PMC11921806 DOI: 10.1083/jcb.202407002] [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: 07/01/2024] [Revised: 12/19/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The mitotic spindle is a bipolar array of microtubules, radiating from the poles which each contain a centrosome, embedded in pericentriolar material. Two proteins, ch-TOG and TACC3, have multiple functions at the mitotic spindle due to operating either alone, together, or in complex with other proteins. To distinguish these activities, we need new molecular tools to dissect their function. Here, we present the structure of the α-helical bundle domain of ch-TOG that mediates its interaction with TACC3 and a structural model describing the interaction, supported by biophysical and biochemical data. We have isolated Affimer tools to precisely target the ch-TOG-binding site on TACC3 in live cells, which displace ch-TOG without affecting the spindle localization of other protein complex components. Inhibition of the TACC3-ch-TOG interaction led unexpectedly to fragmentation of the pericentriolar material in metaphase cells and delayed mitotic progression, uncovering a novel role of TACC3-ch-TOG in maintaining pericentriolar material integrity during mitosis to ensure timely cell division.
Collapse
Affiliation(s)
- James Shelford
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| | - Selena G Burgess
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Elena Rostkova
- School of Cardiovascular and Metabolic Medicine and Sciences and Randall Centre, King's College London, Guy's Campus , London, UK
| | - Mark W Richards
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Gabrielle Larocque
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| | - Josephina Sampson
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Christian Tiede
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Alistair J Fielding
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University , Liverpool, UK
| | - Tina Daviter
- Institute of Cancer Research, Chester Beatty Laboratories , London, UK
| | - Darren C Tomlinson
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Antonio N Calabrese
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Mark Pfuhl
- School of Cardiovascular and Metabolic Medicine and Sciences and Randall Centre, King's College London, Guy's Campus , London, UK
| | - Richard Bayliss
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds , Leeds, UK
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
208
|
Tanaka M, Lum L, Hu KH, Chaudhary P, Hughes S, Ledezma-Soto C, Samad B, Superville D, Ng K, Chumber A, Benson C, Adams ZN, Kersten K, Aguilar OA, Fong L, Combes AJ, Krummel MF, Reeves MQ. Tumor cell heterogeneity drives spatial organization of the intratumoral immune response. J Exp Med 2025; 222:e20242282. [PMID: 40167599 PMCID: PMC11960709 DOI: 10.1084/jem.20242282] [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: 12/02/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Intratumoral heterogeneity (ITH)-defined as genetic and cellular diversity within a tumor-is linked to failure of immunotherapy and an inferior anti-tumor immune response. We modeled heterogeneous tumors comprised of "hot" and "cold" tumor populations (giving rise to T cell-rich and T cell-poor tumors, respectively) and introduced fluorescent labels to enable precise spatial tracking. We found the cold tumor cell population exerted a "dominant cold" effect in mixed tumors. Strikingly, spatial analysis revealed that the tumor cells themselves created distinct local microenvironments within heterogeneous tumors: regions occupied by cold tumor cells showed pronounced immunosuppression, harboring increased CD206Hi macrophages and diminished local T cell function. This inferior T cell activity in cold regions persisted even after immunotherapy and mechanistically was mediated by CX3CL1 produced by the cold tumor cells. An immune cold tumor population within a heterogeneous tumor thus impairs tumor immunity on both a tumor-wide and a highly localized spatial scale.
Collapse
Affiliation(s)
- Miho Tanaka
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Lotus Lum
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth H. Hu
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Piyush Chaudhary
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Savannah Hughes
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Cecilia Ledezma-Soto
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Bushra Samad
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
| | - Daphne Superville
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, USA
| | - Kenneth Ng
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Arun Chumber
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Ciara Benson
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Zoe N. Adams
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Kelly Kersten
- Cancer Metabolism and Microenvironment Program, NCI-designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, LA Jolla, CA, USA
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA, USA
| | - Lawrence Fong
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Alexis J. Combes
- UCSF CoLabs, University of California San Francisco, San Francisco, CA, USA
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Matthew F. Krummel
- ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Melissa Q. Reeves
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
209
|
Ajay AK, Akinfolarin AA, Gifford CC, Sabbisetti VS, Bonventre JV. Breast cancer gene-1 (BRCA1) potentiates maladaptive repair after kidney injury. J Exp Med 2025; 222:e20231107. [PMID: 40152784 PMCID: PMC11951932 DOI: 10.1084/jem.20231107] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2024] [Accepted: 12/06/2024] [Indexed: 03/29/2025] Open
Abstract
Maladaptive repair following kidney tubular injury leads to the development of interstitial fibrosis, a pathology common to chronic kidney diseases (CKD). Dysfunctional DNA damage response plays an important role in the progression of CKD. We found that BRCA1 expression was increased in the kidneys of patients with CKD and fibrotic kidneys of mice. Exon 11 deletion of Brca1 in proximal tubule cells (PTCs) of mice subjected to ischemic or nephrotoxic (aristolochic acid) injury resulted in a reduced number of senescent cells, as assessed by a decrease in phospho-histone H3, p16INK4a, RAD51 recruitment, G2/M cell cycle phase cells, GATA4, and senescence-associated β-galactosidase. There was less production of inflammatory profibrotic mediators and reduced kidney fibrosis. After cisplatin exposure in vitro, human PTCs with reduced BRCA1 had increased apoptosis, decreased RAD51 nuclear foci, and fewer cells in the G2/M cell cycle phase, with reduced IL-6 and sonic hedgehog production. Thus, BRCA1 regulates nonmalignant tissue responses to kidney injury, a role hitherto unrecognized.
Collapse
Affiliation(s)
- Amrendra K. Ajay
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Akinwande A. Akinfolarin
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Cody C. Gifford
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Venkata S. Sabbisetti
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph V. Bonventre
- Renal Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
210
|
Jang G, Park R, Esteva E, Hsu PF, Feng J, Upadhaya S, Sawai CM, Aifantis I, Fooksman DR, Reizis B. Leukemogenic Kras mutation reprograms multipotent progenitors to facilitate its spread through the hematopoietic system. J Exp Med 2025; 222:e20240587. [PMID: 40072317 PMCID: PMC11899982 DOI: 10.1084/jem.20240587] [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: 11/09/2024] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Leukemia-driving mutations are thought to arise in hematopoietic stem cells (HSC), yet the natural history of their spread is poorly understood. We genetically induced mutations within endogenous murine HSC and traced them in unmanipulated animals. In contrast to mutations associated with clonal hematopoiesis (such as Tet2 deletion), the leukemogenic KrasG12D mutation dramatically accelerated HSC contribution to all hematopoietic lineages. The acceleration was mediated by KrasG12D-expressing multipotent progenitors (MPP) that lacked self-renewal but showed increased proliferation and aberrant transcriptome. The deletion of osteopontin, a secreted negative regulator of stem/progenitor cells, delayed the early expansion of mutant progenitors. KrasG12D-carrying cells showed increased CXCR4-driven motility in the bone marrow, and the blockade of CXCR4 reduced the expansion of MPP in vivo. Finally, therapeutic blockade of KRASG12D spared mutant HSC but reduced the expansion of mutant MPP and their mature progeny. Thus, transforming mutations facilitate their own spread from stem cells by reprogramming MPP, creating a preleukemic state via a two-component stem/progenitor circuit.
Collapse
Affiliation(s)
- Geunhyo Jang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rosa Park
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY, USA
| | - Pei-Feng Hsu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Jue Feng
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Samik Upadhaya
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | | | - Iannis Aifantis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - David R. Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
211
|
Lebedev M, Chan FY, Rackles E, Bellessem J, Mikeladze-Dvali T, Xavier Carvalho A, Zanin E. Anillin mediates unilateral furrowing during cytokinesis by limiting RhoA binding to its effectors. J Cell Biol 2025; 224:e202405182. [PMID: 40261302 PMCID: PMC12013513 DOI: 10.1083/jcb.202405182] [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: 05/31/2024] [Revised: 02/10/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
During unilateral furrow ingression, one side of the cytokinetic ring (leading edge) ingresses before the opposite side (lagging edge). Anillin mediates unilateral furrowing during cytokinesis in the one-cell C. elegans zygote by limiting myosin II accumulation in the ring. Here, we address the role of anillin in this process and show that anillin inhibits not only the accumulation of myosin II but also of other RhoA effectors by binding and blocking the RhoA effector site. The interaction between the anillin's RhoA-binding domain (RBD) and active RhoA is enhanced by the disordered linker region and differentially regulated at the leading and lagging edge, which together results in asymmetric RhoA signaling and accumulation of myosin II. In summary, we discover a RhoA GEF- and GAP-independent mechanism, where RhoA activity is limited by anillin binding to the RhoA effector site. Spatial fine-tuning of anillin's inhibitory role on RhoA signaling enables unilateral furrow ingression and contributes to animal development.
Collapse
Affiliation(s)
- Mikhail Lebedev
- Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fung-Yi Chan
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Elisabeth Rackles
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jennifer Bellessem
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| | | | - Ana Xavier Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Esther Zanin
- Department Biologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department Biology II, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|
212
|
Miranda-Alban J, Sanchez-Luege N, Valbuena FM, Rangel C, Rebay I. The Abelson kinase and the Nedd4 family E3 ligases co-regulate Notch trafficking to limit signaling. J Cell Biol 2025; 224:e202407066. [PMID: 40183942 PMCID: PMC11970431 DOI: 10.1083/jcb.202407066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/25/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Precise output from the conserved Notch signaling pathway governs a plethora of cellular processes and developmental transitions. Unlike other pathways that use a cytoplasmic relay, the Notch cell surface receptor transduces signaling directly to the nucleus, with endocytic trafficking providing critical regulatory nodes. Here we report that the cytoplasmic tyrosine kinase Abelson (Abl) facilitates Notch internalization into late endosomes/multivesicular bodies (LEs), thereby limiting signaling output in both ligand-dependent and -independent contexts. Abl phosphorylates the PPxY motif within Notch, a molecular target for its degradation via Nedd4 family ubiquitin ligases. We show that Su(dx), a family member, mediates the Abl-directed LE regulation of Notch via the PPxY, while another family member, Nedd4Lo, contributes to Notch internalization into LEs through both PPxY-dependent and -independent mechanisms. Our findings demonstrate how a network of posttranslational modifiers converging at LEs cooperatively modulates Notch signaling to ensure the precision and robustness of its cellular and developmental functions.
Collapse
Affiliation(s)
- Julio Miranda-Alban
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
| | - Nicelio Sanchez-Luege
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
| | - Fernando M. Valbuena
- Cell and Molecular Biology Graduate Program, University of Chicago, Chicago, IL, USA
| | - Chyan Rangel
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Ilaria Rebay
- Committee on Development, Regeneration, and Stem Cell Biology, University of Chicago, Chicago, IL, USA
- Cell and Molecular Biology Graduate Program, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
213
|
Crncec A, Lau HW, Ng LY, Ma HT, Mak JP, Choi HF, Yeung TK, Poon RYC. Plasticity of mitotic cyclins in promoting the G2-M transition. J Cell Biol 2025; 224:e202409219. [PMID: 40202486 PMCID: PMC11980681 DOI: 10.1083/jcb.202409219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/31/2025] [Accepted: 02/27/2025] [Indexed: 04/10/2025] Open
Abstract
Cyclins and cyclin-dependent kinases (CDKs) orchestrate key events in the cell cycle. However, the uniqueness of individual mitotic cyclins has been a long-standing puzzle. By rapidly removing cyclins in G2 human cells, we found that deficiency of B-type cyclins attenuates mitotic onset and uncouples the G2-M kinase network from mitosis, resulting in sustained activation of PLK1 and cyclin A-CDK1. This culminates in mitotic slippage without completing nuclear envelope breakdown. Remarkably, elevating cyclin A several-fold above its endogenous level is adequate to restore mitosis, allowing cells to survive without B-type cyclins. In contrast, cyclin A is rate-limiting but not essential for G2-M due to compensation by endogenous cyclin B1-CDK2, a non-canonical pair. These findings challenge the traditional indispensable roles of different cyclins and highlight their plasticity. Due to the high malleability of the A- and B-type cyclins, cancer cells may be able to place different weights on different cyclins, while maintaining sufficient CDK activities for successful mitosis.
Collapse
Affiliation(s)
- Adrijana Crncec
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ho Wai Lau
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Lau Yan Ng
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hoi Tang Ma
- Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Joyce P.Y. Mak
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Hon Fung Choi
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Tsz Kwan Yeung
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Yat Choi Poon
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
214
|
Isogai T, Hirosawa KM, Kanno M, Sho A, Kasai RS, Komura N, Ando H, Furukawa K, Ohmi Y, Furukawa K, Yokota Y, Suzuki KG. Extracellular vesicles adhere to cells primarily by interactions of integrins and GM1 with laminin. J Cell Biol 2025; 224:e202404064. [PMID: 40304687 PMCID: PMC12042775 DOI: 10.1083/jcb.202404064] [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: 04/12/2024] [Revised: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 05/02/2025] Open
Abstract
Tumor-derived extracellular vesicles (EVs) have attracted significant attention, yet the molecular mechanisms that govern their specific binding to recipient cells remain elusive. Our in vitro study utilizing single-particle tracking demonstrated that integrin heterodimers comprising α6β4 and α6β1 and ganglioside, GM1, are responsible for the binding of small EV (sEV) subtypes to laminin. EVs derived from four distinct tumor cell lines, regardless of size, exhibited high binding affinities for laminin but not for fibronectin, although fibronectin receptors are abundant in EVs and have functional roles in EV-secreting cells. Our findings revealed that integrins in EVs bind to laminin via the conventional molecular interface, facilitated by CD151 rather than by inside-out signaling of talin-1 and kindlin-2. Super-resolution movie observation revealed that sEV integrins bind only to laminin on living recipient cells. Furthermore, sEVs bound to HUVEC and induced cell branching morphogenesis in a laminin-dependent manner. Thus, we demonstrated that EVs predominantly bind to laminin on recipient cells, which is indispensable for cell responses.
Collapse
Affiliation(s)
- Tatsuki Isogai
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
| | | | - Miki Kanno
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
| | - Ayano Sho
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Rinshi S. Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Hiromune Ando
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Keiko Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yuhsuke Ohmi
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Koichi Furukawa
- Department of Biomedical Sciences, Chubu University, Kasugai, Japan
| | - Yasunari Yokota
- Department of Information Science, Faculty of Engineering, Gifu University, Gifu, Japan
| | - Kenichi G.N. Suzuki
- The United Graduate School of Agricultural Science, Gifu University, Gifu, Japan
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
- Graduate School of Natural Science and Technology, Gifu University, Gifu, Japan
- Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, Japan
- Innovation Research Center for Quantum Medicine, Graduate School of Medicine, Gifu University, Gifu, Japan
| |
Collapse
|
215
|
Ptok J, Theiss S, Schaal H. Fully haplotyped genome assemblies of healthy individuals reveal variability in 5'ss strength and support by splicing regulatory proteins. NAR Genom Bioinform 2025; 7:lqaf036. [PMID: 40191587 PMCID: PMC11970367 DOI: 10.1093/nargab/lqaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/18/2025] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
This work presents a comprehensive investigation of sequence variations at human 5' splice sites (5'ss), exploring their impact on 5'ss strength and predicted splicing regulatory protein (SRP) binding. Leveraging 44 high-quality genomes, with fully haplotyped assemblies, we were able to fully assess homozygous and heterozygous sequence variations around and within 5'ss. Variations showed differing tolerance levels in protein-coding and non-coding transcripts. Around half of 5'ss variations did not alter 5'ss strength (measured by the HBond score, that estimates binding of the 11 nucleotides at the free U1 spliceosomal RNA 5' end). Heterozygous variations resulted in stronger 5'ss strength reductions and less compensatory effects of multiple sequence variations at the same 5'ss, compared to homozygous variations. Additionally, we observed a slight balance between changes in 5'ss strength and predicted SRP binding. Strong 5'ss (HBond score >18.8) showed strength variations in both directions, as theoretically expected for random distributions, whereas weaker 5'ss consistently showed lower strength reductions than expected or achievable. Variations at 5'ss of essential genes were less frequent than in other genes and showed a higher amount of variations that did not alter 5'ss strength. Acceptable changes in predicted SRP binding sites were highly dependent on their respective 5'ss strength.
Collapse
Affiliation(s)
- Johannes Ptok
- Institute of Virology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Stephan Theiss
- Institute of Virology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Heiner Schaal
- Institute of Virology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| |
Collapse
|
216
|
Hurrah IM, Mohammad, Kumar A, Abbas N. Synergistic interaction of AaMYC2 and AaMYC2-LIKE enhances artemisinin production in Artemisia annua L. J Biotechnol 2025; 402:69-78. [PMID: 40107365 DOI: 10.1016/j.jbiotec.2025.03.007] [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: 12/04/2024] [Revised: 02/07/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
Artemisinin-based combination therapies recommended by WHO marks Artemisia annua as the only natural source of artemisinin fighting deadly disease, Malaria. In this study, we isolated two transcription factors, AaMYC2 and AaMYC2-LIKE, from A. annua and investigated their role in regulating artemisinin biosynthetic pathway. Our findings depict that both AaMYC2 and AaMYC2-LIKE are transcriptionally active and, when co-transformed in yeast cells, significantly enhance β-galactosidase activity in transactivation assays as compared to their individual transformations. Furthermore, Yeast two-hybrid (Y2H) and Biomolecular fluorescence complementation assays revealed AaMYC2 physically interacts with AaMYC2-LIKE in yeast cells and in the nucleus of onion epidermal cells respectively. Generation of transient transgenic over expression and co-expression lines of AaMYC2 and AaMYC2-LIKE resulted in elevated expression of artemisinin biosynthetic genes and trichome development genes in co-expression lines as compared to individual transgenic lines and wildtype. Importantly, the glandular trichome density and artemisinin content was also significantly higher in co-transformed transgenic lines compared to individual AaMYC2 and AMYC2-LIKE transgenic lines. Conversely, artemisinin content was markedly reduced in AaMYC2-RNAi lines, underscoring the critical role of functional AaMYC2 in synergistic regulation with AaMYC2-LIKE. Altogether the above studies provide valuable insights into the regulatory networks of MYC type bHLH transcription factors in controlling economically and medically important pathway in A. annua.
Collapse
Affiliation(s)
- Ishfaq Majid Hurrah
- Plant Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, J&K 190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India
| | - Mohammad
- Plant Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, J&K 190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India
| | - Amit Kumar
- Instrumentation Division, CSIR-Indian Institute of Integrative Medicine, Jammu Tawi, 180001, India
| | - Nazia Abbas
- Plant Biotechnology Division, CSIR-Indian Institute of Integrative Medicine, Sanat Nagar, Srinagar, J&K 190005, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, UP 201002, India.
| |
Collapse
|
217
|
Márquez-Mendoza JM, Baranda-Ávila N, Lizano M, Langley E. Micro-RNAs targeting the estrogen receptor alpha involved in endocrine therapy resistance in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167783. [PMID: 40057206 DOI: 10.1016/j.bbadis.2025.167783] [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: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Endocrine therapy resistance (ETR) in breast cancer (BC) is a multicausal phenomenon with diverse alterations in the tumor cell interactome. Within these alterations, non-coding RNAs (ncRNAs) such as micro-RNAs (miRNAs) modulate the expression of tumor suppressor genes and proto-oncogenes, such as the ESR1 gene encoding estrogen receptor alpha (ERα). This work aims to review the effects of miRNAs targeting ERα mRNA and their mechanisms related to ETR in BC. A thorough review of the literature and an in silico study were carried out to elucidate the involvement of each miRNA, thus contributing to the understanding of ETR in BC.
Collapse
Affiliation(s)
- J M Márquez-Mendoza
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - N Baranda-Ávila
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - M Lizano
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - E Langley
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| |
Collapse
|
218
|
Elshaer N, Escudero J, Piulachs MD. The transcription factor Capicua maintains the oocyte polarity in the panoistic ovary of the German cockroach. Dev Biol 2025; 522:125-135. [PMID: 40158791 DOI: 10.1016/j.ydbio.2025.03.016] [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: 11/04/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
The establishment of the symmetry axis is crucial for the development of all organisms. In insects, this process begins early in oogenesis with the correct distribution of the mRNAs and proteins in the oocyte. One protein that plays a role in organizing this distribution is the transcription factor Capicua (Cic). Cic has been studied in the context of oogenesis and embryonic development in Drosophila melanogaster. It is maternally expressed, begins essential for establishing the dorsoventral axis, and functions as a transcriptional repressor. Although the Cic sequences are conserved across species, their function in other types of insect ovaries is still little known. We wondered whether the function of Cic in insects has been maintained through evolution despite the ovary type or if it has been modified in parallel to the ovary evolution. To address this, we studied the Cic function in a phylogenetically basal insect, the cockroach Blattella germanica, a species with panoistic ovaries. Our findings show that B. germanica Cic is essential for oocyte development and the maturation of ovarian follicles. A loss of Cic function leads to disrupted cytoskeletal organization, defects in anterior-posterior polarity, and compromised follicle integrity. The conservation and functional divergence of Cic across different species suggest evolutionary adaptations in the mechanisms of insect oogenesis.
Collapse
Affiliation(s)
- Nashwa Elshaer
- Institute of Evolutionary Biology (CSIC-University Pompeu Fabra), Passeig Marítim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - Jorge Escudero
- Institute of Evolutionary Biology (CSIC-University Pompeu Fabra), Passeig Marítim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - Maria-Dolors Piulachs
- Institute of Evolutionary Biology (CSIC-University Pompeu Fabra), Passeig Marítim de la Barceloneta, 37, 08003, Barcelona, Spain.
| |
Collapse
|
219
|
Sipani R, Rawal Y, Barman J, Abburi P, Kurlawala V, Joshi R. Drosophila grainyhead gene and its neural stem cell specific enhancers show epigenetic synchrony in the cells of the central nervous system. Dev Biol 2025; 522:227-239. [PMID: 40154783 DOI: 10.1016/j.ydbio.2025.03.012] [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: 12/23/2024] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
Enhancers are the epicentres of tissue-specific gene regulation. In this study, we have used the central nervous system (CNS) specific expression of the Drosophila grainyhead (grh) gene to make a case for deleting the enhancers in a sensitised background of other enhancer deletion, to functionally validate their role in tissue-specific gene regulation. We identified novel enhancers for grh and subsequently deleted two of them, to establish their collective importance in regulating grh expression in CNS. This showed that grh relies on multiple enhancers for its robust expression in neural stem cells (NSCs), with different combinations of enhancers playing a critical role in regulating its expression in various subset of these cells. We also found that these enhancers and the grh gene show epigenetic synchrony across the three cell types (NSCs, intermediate progenitors and neurons) of the developing CNS; and grh is not transcribed in intermediate progenitor cells, which inherits the Grh protein from the NSCs. We propose that this could be a general mechanism for regulating the expression of cell fate determinant protein in intermediate progenitor cells. Lastly, our results underline that enhancer redundancy results in phenotypic robustness in grh gene expression, which seems to be a consequence of the cumulative activity of multiple enhancers.
Collapse
Affiliation(s)
- Rashmi Sipani
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India; Manipal Academy of Higher Education, Manipal, 576104, India
| | - Yamini Rawal
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India; Regional Centre for Biotechnology, Faridabad, 121001, India
| | - Jiban Barman
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India; Manipal Academy of Higher Education, Manipal, 576104, India
| | - Prakeerthi Abburi
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India
| | - Vishakha Kurlawala
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India
| | - Rohit Joshi
- Laboratory of Neuroscience and Cell Biology, Centre for DNA Fingerprinting and Diagnostics (CDFD), BRIC-CDFD, Inner Ring Road, Uppal, Hyderabad, 500039, India.
| |
Collapse
|
220
|
Liu M, Fang T, Wang S, Ma H, Kong L, Deng X, Teng Z, Wang J, Zhang P, Xu L. Repurposing tavaborole to combat resistant bacterial infections through competitive inhibition of KPC-2 and metabolic disruption. Bioorg Chem 2025; 159:108421. [PMID: 40179579 DOI: 10.1016/j.bioorg.2025.108421] [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: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
The rise of carbapenem-resistant Enterobacteriaceae (CRE) strains has emerged as an increasing threat to global public health. The development of antibiotic adjuvants presents an economical and promising approach to address this crisis. Through a high-throughput screen of the FDA-approved compound library, we identified tavaborole (AN2690) as a broad-spectrum β-lactamase inhibitor. The mechanistic study revealed that tavaborole formed a reversible binding with the active serine of KPC-2, showing effective competitive inhibition. Its electron-deficient boron atom formed a borate ester bond with hydroxyl group of the serine residue at the active site of KPC-2, transitioning to an sp3-hybridized state that mimicked the tetrahedral intermediate during KPC-2 catalytic. Moreover, transcriptomic analysis and bacterial metabolism assays further unveiled tavaborole addition can inhibit tricarboxylic acid (TCA) cycle, coupled with downregulation of intracellular ATP levels, indicating that tavaborole compromised the bacterial metabolic homeostasis and exerted synergistic antibacterial activity. Notably, the combination treatment further suppressed the development of meropenem resistance. In mouse intraperitoneal infection models, tavaborole effectively restored the efficacy of meropenem against CRE bacteria. These findings elucidate the synergistic mechanisms of tavaborole, expand its potential applications in anti-infection therapeutics, and provide a promising strategy for addressing CRE infections.
Collapse
Affiliation(s)
- Minda Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; 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 130062, China
| | - Tianqi Fang
- 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 130062, China; Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Shanshan Wang
- 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 130062, China
| | - Hongxia Ma
- College of Animal Science and Technology, Jilin Agricultural University, The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, China
| | - Lingcong Kong
- College of Animal Science and Technology, Jilin Agricultural University, The Key Laboratory of New Veterinary Drug Research and Development of Jilin Province, Jilin Agricultural University, Changchun 130118, China
| | - Xuming Deng
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; 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 130062, China
| | - Zihao Teng
- 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 130062, China
| | - Jianfeng Wang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; 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 130062, China
| | - Peng Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China.
| | - Lei Xu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China; 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 130062, China.
| |
Collapse
|
221
|
Sun C, Ding Z, Li B, Chen S, Li E, Yang Q. New insights into Gremlin-1: A tumour microenvironment landscape re-engineer and potential therapeutic target. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119962. [PMID: 40250712 DOI: 10.1016/j.bbamcr.2025.119962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Gremlin-1 (GREM1), a well-known bone morphogenetic protein (BMP) antagonist, is highly expressed in various malignant tumours. However, the specific role of GREM1 in tumours remains controversial and may be attributed to the heterogeneity and complexity of the tumour microenvironment (TME). It is currently believed that GREM1 regulates the complex landscape of the TME, primarily by antagonising BMP signalling or BMP-independent pathways. Both GREM1 and BMP play dual roles in tumour progression. Therefore, the mutual crosstalk between tumour cells and tumour-associated fibroblasts and the regulation of various secreted factors in the TME affect the secretion level of GREM1, which in turn regulates the amplitude balance between GREM1 and BMP, affecting tumour progression. The inhibition of GREM1 activity in the TME can disrupt this amplitude balance and prevent the formation of a tumour-supportive microenvironment, demonstrating that GREM1 is a potential therapeutic target. In this study, we reviewed the specific signalling pathways via which GREM1 in the TME regulates epithelial-mesenchymal transition, construction of the tumour immune microenvironment, and maintenance of tumour cell stemness via BMP-dependent and BMP-independent regulation, and also summarised the latest clinical progress of GREM1.
Collapse
Affiliation(s)
- Chengpeng Sun
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang 330006, China; HuanKui Academy, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Zijun Ding
- School of Ophthalmology and Optometry, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Benjie Li
- Queen Mary School, Jiangxi Medical college, Nanchang University, Nanchang 330031, China
| | - Sihong Chen
- Queen Mary School, Jiangxi Medical college, Nanchang University, Nanchang 330031, China
| | - Enliang Li
- Department of General Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang 330006, China; Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, China.
| | - Qingping Yang
- Department of Reproductive Medicine, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17, Yongwai zheng Street, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
222
|
Malin SK. Circadian rhythms and gastrointestinal hormone-related appetite regulation. Curr Opin Endocrinol Diabetes Obes 2025; 32:97-101. [PMID: 40110812 PMCID: PMC12043425 DOI: 10.1097/med.0000000000000908] [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] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW Circadian biology influences the gastrointestinal system as exemplified by hormonal patterns that modulate appetite. Indeed, people tend to get hungrier towards the later parts of the day. How misalignment of our circadian biology with behavioral factors (i.e. diet, exercise, sleep, etc.) influences obesity related disease has been an area of intense recent investigation. RECENT FINDINGS The gastrointestinal hormones (e.g. ghrelin, glucagon-like polypeptide-1, glucose dependent insulinotrophic peptide, peptide tyrosine-tyrosine, and insulin) play unique roles across the 24-h cycle in fostering anticipatory responses that promote desires to eat while concurrently responding to environmental stimuli. A persons chronotype has emerged as a target area since it provides a metric of circadian biology interacting with environmental factors and affects all people. In fact, later chronotypes tend to be at higher risk for obesity, due to in part, alterations in gastrointestinal hormones (e.g. GIP, insulin) that align with behavioral observations of greater food intake and desires to eat fatty/sweet foods later in the day. SUMMARY Changes in gastrointestinal hormones across the 24-h cycle impact obesity risk when misalignment of our circadian biology occurs with behavioral cycles. Better understanding how chronotype modulates appetite may enable personalized prescription of exercise, diet and/or medication to foster reduced chronic disease risk.
Collapse
Affiliation(s)
- Steven K. Malin
- Department of Kinesiology & Health, Rutgers University, New Brunswick, NJ
- Division of Endocrinology, Metabolism & Nutrition; Rutgers University, New Brunswick, NJ
- New Jersey Institute for Food, Nutrition and Health, Rutgers University, New Brunswick, NJ
- Institute of Translational Medicine and Science, Rutgers University, New Brunswick, NJ
| |
Collapse
|
223
|
Gong L, Wu L, Zhao S, Xiao S, Chu X, Zhang Y, Li F, Li S, Yang H, Jiang P. Epigenetic regulation of ferroptosis in gastrointestinal cancers (Review). Int J Mol Med 2025; 55:93. [PMID: 40242977 PMCID: PMC12045471 DOI: 10.3892/ijmm.2025.5534] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Ferroptosis is a type of iron‑dependent cell death characterized by excessive lipid peroxidation and may serve as a potential therapeutic target in cancer treatment. While the mechanisms governing ferroptosis continue to be explored and elucidated, an increasing body of research highlights the significant impact of epigenetic modifications on the sensitivity of cancer cells to ferroptosis. Epigenetic processes, such as DNA methylation, histone modifications and non‑coding RNAs, have been identified as key regulators that modulate the expression of ferroptosis‑related genes. These alterations can either enhance or inhibit the sensitivity of gastrointestinal cancer (GIC) cells to ferroptosis, thereby affecting the fate of GICs. Drugs that target epigenetic markers for advanced‑stage cancer have shown promising results in enhancing ferroptosis and inhibiting tumor growth. This review explores the intricate relationship between epigenetic regulation and ferroptosis in GICs. Additionally, the potential of leveraging epigenetic modifications to trigger ferroptosis in GICs is investigated. This review highlights the importance of further research to elucidate the specific mechanisms underlying epigenetic control of ferroptosis and to advance the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Linlin Wu
- Oncology Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| | - Shuai Xiao
- Department of Intensive Care Medicine, Tengzhou Central People's Hospital, Jining Medical University, Tengzhou, Shandong 277500, P.R. China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
| | - Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Fengfeng Li
- Neurosurgery Department, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuhui Li
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Hui Yang
- Department of Gynecology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People's Hospital, Shandong First Medical University, Jining, Shandong 272000, P.R. China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, Shandong 272000, P.R. China
| |
Collapse
|
224
|
Ukita N, Ogawa T, Yamada M, Takeuchi C, Kosaki K, Moriyama K. Functional Analyses of SATB2 Variants Reveal Pathogenicity Mechanisms Linked With SATB2-Associated Syndrome. Am J Med Genet A 2025; 197:e64005. [PMID: 39887889 DOI: 10.1002/ajmg.a.64005] [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: 10/24/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
SATB2-associated syndrome (SAS) is characterized by intellectual disability, neurodevelopmental disorders, cleft palate, and dental abnormalities. SAS is caused by variants in the special AT-rich sequence-binding protein 2 (SATB2), which encodes a transcription factor containing two CUT domains and a homeobox (HOX) domain. Here, we report the case of a 16-year-old male diagnosed with SAS using exome sequencing and investigate the functional consequences of previously reported SATB2 variants, including those in this case. The patient carried a heterozygous missense variant (c.1147G>C, p.A383P) in SATB2, which was predicted to be pathogenic in silico but was absent from public databases. Immunofluorescence assays demonstrated that SATB2 proteins with variants in the CUT2 domain predominantly localized to the cytoplasm. Functional analysis further revealed that wild-type SATB2 increased the activity of the Msx1 promoter, which is involved in palatogenesis and tooth development, whereas variants in the CUT1 domain disrupted this transcriptional activation. These findings suggest that the nuclear localization signal of SATB2 resides in the CUT2 domain and that Msx1 promoter impairment owing to SATB2 variants may contribute to the pathogenesis of cleft palate and tooth agenesis in SAS. This research highlights a novel pathogenic variant and the functional implications for understanding SAS.
Collapse
Affiliation(s)
- Nao Ukita
- Department of Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Takuya Ogawa
- Department of Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Mamiko Yamada
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Chisen Takeuchi
- Department of Clinical Genetics, Jikei University Hospital, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
225
|
Tinant G, Neefs I, De Groote A, Page MM, Rees JF, Larondelle Y, Debier C. Docosapentaenoic (22:5 n-6) and docosahexaenoic (22:6 n-3) acids exhibit highly lipogenic properties in rainbow trout preadipocytes. Comp Biochem Physiol B Biochem Mol Biol 2025; 278:111087. [PMID: 39993557 DOI: 10.1016/j.cbpb.2025.111087] [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: 11/20/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Dietary polyunsaturated fatty acids are essential for fish health. Adipose tissue is the major tissue for fatty acid storage in rainbow trout (Oncorhynchus mykiss), and its development and function can be impacted by the fatty acids themselves. In the present study, the effects of seven fatty acids, oleic (OA, 18:1 n-9), α-linolenic (ALA, 18:3 n-3), eicosapentaenoic (EPA, 20:5 n-3), docosahexaenoic (DHA, 22:6 n-3), linoleic (LA, 18:2 n-6), arachidonic (AA, 20:4 n-6), and docosapentaenoic (DPA, 22:5 n-6) acids, on adipogenesis were investigated in primary cultures of rainbow trout preadipocytes. In terms of lipid accumulation, DPA and DHA appeared to be the most lipogenic fatty acids, while all treatments modified the fatty acid composition of the cellular phospholipids and neutral lipids. The fatty acid of interest added to the culture medium was the most abundant in preadipocytes, while the first bioconversion products were detected in lower amounts. In terms of transcriptional effects, DPA increased the expression of the early transcription factor CCAAT/enhancer binding protein δ, while DHA upregulated the expression of genes involved in neutral lipid synthesis, notably lipoprotein lipase, fatty acid transport protein 1 and glycerol-3-phosphate dehydrogenase. Both fatty acids decreased the expression of fatty acid synthase. These results highlight that DPA and DHA exert a significant effect on lipid deposition in rainbow trout preadipocytes, potentially through different pathways, and confirm that fatty acids have major impacts on preadipocyte lipid metabolism and adipogenesis.
Collapse
Affiliation(s)
- Gilles Tinant
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium.
| | - Ineke Neefs
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium
| | - Alice De Groote
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium
| | - Melissa M Page
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium
| | - Jean-François Rees
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium
| | - Yvan Larondelle
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium
| | - Cathy Debier
- Louvain Institute of Biomolecular Science and Technology (LIBST), Université catholique de Louvain, Croix du Sud 4-5/L7.07.03, 1348 Louvain-la-Neuve, Belgium.
| |
Collapse
|
226
|
Honjo Y, Ichinohe T. Neural crest cells are sensitive to radiation-induced DNA damage. Tissue Cell 2025; 94:102774. [PMID: 39954562 DOI: 10.1016/j.tice.2025.102774] [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: 07/19/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Radiation-induced DNA damage introduces mutations that have various deleterious effects, which may lead to apoptosis and carcinogenesis. Different tissues and cell types exhibit varying degrees of sensitivity to radiation-induced DNA damage, which is often attributed to the frequency of cell division. In this study, we showed that irradiation affects early zebrafish embryos in a manner that is not explained by direct DNA damage and repair nor by the frequency of cell division. Zebrafish embryos irradiated at 2 h post fertilization showed drastic apoptosis, mainly in the head region, during organogenesis. Herein, we show that these apoptotic cells did not show aneuploidy or micronuclei, and that not all descendants of the same cells with the same DNA damage were necessarily apoptotic. Finally, we demonstrate that apoptotic cells have various origins and that neural crest cells have a sensitive cell fate. Our results suggest the existence of a radiation damage response mechanism other than those previously described, the elucidation of which may inform strategies for greater protection against radiation injury.
Collapse
Affiliation(s)
- Yasuko Honjo
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima 734-8553, Japan.
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine (RIRBM), Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
227
|
Alsaedi S, Ogasawara M, Alarawi M, Gao X, Gojobori T. AI-powered precision medicine: utilizing genetic risk factor optimization to revolutionize healthcare. NAR Genom Bioinform 2025; 7:lqaf038. [PMID: 40330081 PMCID: PMC12051108 DOI: 10.1093/nargab/lqaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/11/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
The convergence of artificial intelligence (AI) and biomedical data is transforming precision medicine by enabling the use of genetic risk factors (GRFs) for customized healthcare services based on individual needs. Although GRFs play an essential role in disease susceptibility, progression, and therapeutic outcomes, a gap exists in exploring their contribution to AI-powered precision medicine. This paper addresses this need by investigating the significance and potential of utilizing GRFs with AI in the medical field. We examine their applications, particularly emphasizing their impact on disease prediction, treatment personalization, and overall healthcare improvement. This review explores the application of AI algorithms to optimize the use of GRFs, aiming to advance precision medicine in disease screening, patient stratification, drug discovery, and understanding disease mechanisms. Through a variety of case studies and examples, we demonstrate the potential of incorporating GRFs facilitated by AI into medical practice, resulting in more precise diagnoses, targeted therapies, and improved patient outcomes. This review underscores the potential of GRFs, empowered by AI, to enhance precision medicine by improving diagnostic accuracy, treatment precision, and individualized healthcare solutions.
Collapse
Affiliation(s)
- Sakhaa Alsaedi
- Computer Science, Division of Computer, Electrical and Mathematical Sciences and Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- College of Computer Science and Engineering (CCSE), Taibah University, 42353 Madinah, Kingdom of Saudi Arabia
| | - Michihiro Ogasawara
- Department of Internal Medicine and Rheumatology, Juntendo University, 113-8431 Tokyo, Japan
| | - Mohammed Alarawi
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
| | - Xin Gao
- Computer Science, Division of Computer, Electrical and Mathematical Sciences and Engineering (CEMSE), King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
| | - Takashi Gojobori
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), 23955-6900 Thuwal, Kingdom of Saudi Arabia
- Marine Open Innovation Institute (MaOI), 113-8431 Shizuoka, Japan
| |
Collapse
|
228
|
Wang C, Bai J, Xiong H, Xie Y, Gu J, Zhao L, Li H, Ding Y, Guo X, Guo H, Liu L. Characterization of wheat oligo-tiller mutant ot2 and fine mapping of the mutant gene Taot2. THE PLANT GENOME 2025; 18:e70043. [PMID: 40341859 DOI: 10.1002/tpg2.70043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/25/2025] [Accepted: 04/06/2025] [Indexed: 05/11/2025]
Abstract
Tiller number is a crucial determinant of grain yield in wheat (Triticum aestivum L.), and identifying functional alleles can enhance our understanding of wheat tiller development. Here, we describe the ot2 wheat mutant, which exhibits a 91% reduction in tiller number compared to the wild type and displays inhibited tiller bud differentiation from the 2-3 leaf stage. Bulked segregant analysis combined with exon sequencing mapped the Taot2 gene to chromosome 1BL. Fourteen kompetitive allele-specific PCR markers were developed and utilized to narrow down the region containing Taot2 to a 2.22 Mb interval. This region encompasses 39 high-confidence genes. Through transcriptomic analysis and functional gene investigation, TraesCS1B03G1126300 was identified as the candidate gene, encoding an auxin-responsive protein of the auxin/indole-3-acetic acid family. Our findings provide the foundation for map-based cloning of Taot2, a novel wheat tiller gene, and offer valuable insights into wheat tiller bud initiation.
Collapse
Affiliation(s)
- Chenxi Wang
- College of Life Sciences, Key Laboratory of Major Crop Germplasm Innovation and Application in Qingdao, Qingdao Agricultural University, Qingdao, China
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiaxing Bai
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongchun Xiong
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yongdun Xie
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jiayu Gu
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Linshu Zhao
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Huiyuan Li
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuping Ding
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xinmei Guo
- College of Life Sciences, Key Laboratory of Major Crop Germplasm Innovation and Application in Qingdao, Qingdao Agricultural University, Qingdao, China
| | - Huijun Guo
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Luxiang Liu
- State Key Laboratory of Crop Gene Resources and Breeding/National Engineering Laboratory of Crop Molecular Breeding/CAEA Research and Development Centre on Nuclear Technology Applications for Irradiation Mutation Breeding, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
229
|
Saberiyan M, Gholami S, Ejlalidiz M, Rezaeian Manshadi M, Noorabadi P, Hamblin MR. The dual role of chaperone-mediated autophagy in the response and resistance to cancer immunotherapy. Crit Rev Oncol Hematol 2025; 210:104700. [PMID: 40086769 DOI: 10.1016/j.critrevonc.2025.104700] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy has become a revolutionary strategy in oncology, utilizing the host immune system to fight malignancies. Notwithstanding major progress, obstacles such as immune evasion by tumors and the development of resistance still remain. This manuscript examines the function of chaperone-mediated autophagy (CMA) in cancer biology, focusing on its effects on tumor immunotherapy response and resistance. CMA is a selective degradation mechanism for cytosolic proteins, which is crucial for sustaining cellular homeostasis and regulating immune responses. By degrading specific proteins, CMA can either facilitate tumor progression in stressful conditions, or promote tumor suppression by removing oncogenic factors. This double-edged sword highlights the complexity of CMA in cancer progression and its possible effect on treatment results. Here we clarify the molecular mechanisms by which CMA can regulate the immune response and its possible role as a therapeutic target for improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sarah Gholami
- Young Researchers and Ellie Club, Babol Branch. Islamic Azad University, Babol, Iran
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical sciences, Urmia, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
230
|
Khalife M, Jia T, Caron P, Shreim A, Genoux A, Cristini A, Pucciarelli A, Leverve M, Lepeltier N, García-Rodríguez N, Dalonneau F, Ramachandran S, Fernandez Martinez L, Marcion G, Lemaitre N, Brambilla E, Garrido C, Hammond E, Huertas P, Gazzeri S, Sordet O, Eymin B. SRSF2 overexpression induces transcription-/replication-dependent DNA double-strand breaks and interferes with DNA repair pathways to promote lung tumor progression. NAR Cancer 2025; 7:zcaf011. [PMID: 40181846 PMCID: PMC11963763 DOI: 10.1093/narcan/zcaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 02/04/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
SRSF2 (serine/arginine-rich splicing factor 2) is a critical regulator of pre-messenger RNA splicing, which also plays noncanonical functions in transcription initiation and elongation. Although elevated levels of SRSF2 are associated with advanced stages of lung adenocarcinoma (LUAD), the mechanisms connecting SRSF2 to lung tumor progression remain unknown. We show that SRSF2 overexpression increases global transcription and replicative stress in LUAD cells, which correlates with the production of DNA damage, notably double-strand breaks (DSBs), likely resulting from conflicts between transcription and replication. Moreover, SRSF2 regulates DNA repair pathways by promoting homologous recombination and inhibiting nonhomologous end joining. Mechanistically, SRSF2 interacts with and enhances MRE11 (meiotic recombination 11) recruitment to chromatin, while downregulating 53BP1 messenger RNA and protein levels. Both events are likely contributing to SRSF2-mediated DNA repair process rerouting. Lastly, we show that SRSF2 and MRE11 expression is commonly elevated in LUAD and predicts poor outcome of patients. Altogether, our results identify a mechanism by which SRSF2 overexpression promotes lung cancer progression through a fine control of both DSB production and repair. Finally, we show that SRSF2 knockdown impairs late repair of ionizing radiation-induced DSBs, suggesting a more global function of SRSF2 in DSB repair by homologous recombination.
Collapse
Affiliation(s)
- Manal Khalife
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Tao Jia
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Pierre Caron
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Amani Shreim
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Aurelie Genoux
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Agnese Cristini
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Marie Leverve
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Nina Lepeltier
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Néstor García-Rodríguez
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Fabien Dalonneau
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Shaliny Ramachandran
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Lara Fernandez Martinez
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Guillaume Marcion
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Nicolas Lemaitre
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Elisabeth Brambilla
- University Grenoble Alpes, INSERM U1209, CNRS UMR 5309, Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Carmen Garrido
- INSERM, UMR1231, Faculty of Medicine and Pharmacy, Université de Bourgogne Franche-Comté, Dijon F21000, France
| | - Ester M Hammond
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Pablo Huertas
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla 41080, Spain; Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad de Sevilla/CSIC, Sevilla 41092, Spain
| | - Sylvie Gazzeri
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| | - Olivier Sordet
- Cancer Research Center of Toulouse (CRCT), INSERM, Université de Toulouse, CNRS, Toulouse 31037, France
| | - Beatrice Eymin
- University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Team RNA Splicing, Cell Signaling and Response to Therapies, Institute for Advanced Biosciences, Grenoble F38000, France
| |
Collapse
|
231
|
Alborzi N, Maroofi A, Hafizi Barjin Z, Moradi A, Rezvani ME, Safari F. Resveratrol attenuates pressure overload-induced myocardial remodeling in ovariectomized rats by rescuing the adaptive angiogenic response. Life Sci 2025; 370:123573. [PMID: 40122333 DOI: 10.1016/j.lfs.2025.123573] [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: 01/03/2025] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Resveratrol (RES), a polyphenol with putative estrogen (E2) -like effects, is believed to counteract left ventricular hypertrophy (LVH). However, how RES exerts its protection is not well understood, particularly when prominent risk factors, such as E2 depletion and pressure overload (PO), coexist. Here, we evaluated the impact of RES and E2 on angiogenesis and LVH in rats subjected to ovariectomy (OVX) and PO. METHODS Three weeks after bilateral OVX induction, abdominal aortic banding was performed on Wistar female rats to trigger PO. The animals were treated with either RES or E2 for six weeks. Finally, the heart-to-body weight ratio (HW/BW), cell size, fibrosis, and atrial natriuretic peptide (ANP) mRNA expression were assessed. Angiogenesis was determined by evaluating vascular endothelial growth factor (VEGF) mRNA and protein expression and by CD31 immunostaining. Serum E2 levels were also measured. RESULTS OVX + PO caused more severe myocardial hypertrophy (HW/BW) and fibrosis compared with PO alone, but did not aggravate cell size and ANP mRNA expression. OVX blunted the angiogenic response to PO, with reduced VEGF expression. RES increased VEGF expression and CD31, and abrogated LVH and fibrosis. E2 treatment improved VEGF expression and fibrosis, but not to the same extent as RES. RES improved serum levels of E2 in OVX + PO rats. CONCLUSION Our findings suggest that RES limits OVX-induced exacerbation of LVH and fibrosis in a PO model, and targets systemic E2 levels and myocardial angiogenesis as underpinning protective mechanisms. Thus, RES may provide cardioprotection for post-menopausal women.
Collapse
Affiliation(s)
- Nasrin Alborzi
- Yazd Neuroendocrine Research Center, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Zeinab Hafizi Barjin
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Moradi
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ebrahim Rezvani
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Safari
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
232
|
Yanase R, Zeeshan M, Ferguson DJ, Markus R, Brady D, Bottrill AR, Holder AA, Guttery DS, Tewari R. Divergent Plasmodium kinases drive MTOC, kinetochore and axoneme organisation in male gametogenesis. Life Sci Alliance 2025; 8:e202403056. [PMID: 40127922 PMCID: PMC11933671 DOI: 10.26508/lsa.202403056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
Sexual development and male gamete formation of the malaria parasite in the mosquito midgut are initiated by rapid endomitosis in the activated male gametocyte. This process is highly regulated by protein phosphorylation, specifically by three divergent male-specific protein kinases (PKs): CDPK4, SRPK1, and MAP2. Here, we localise each PK during male gamete formation using live-cell imaging, identify their putative interacting partners by immunoprecipitation, and determine the morphological consequences of their absence using ultrastructure expansion and transmission electron microscopy. Each PK has a distinct location in either the nuclear or the cytoplasmic compartment. Protein interaction studies revealed that CDPK4 and MAP2 interact with key drivers of rapid DNA replication, whereas SRPK1 is involved in RNA translation. The absence of each PK results in severe defects in either microtubule-organising centre organisation, kinetochore segregation, or axoneme formation. This study reveals the crucial role of these PKs during endomitosis in formation of the flagellated male gamete and uncovers some of their interacting partners that may drive this process.
Collapse
Affiliation(s)
- Ryuji Yanase
- School of Life Sciences, University of Nottingham, Nottingham, UK
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK
| | - Mohammad Zeeshan
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - David Jp Ferguson
- Nuffield Department of Clinical Laboratory Sciences and John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| | - Robert Markus
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Declan Brady
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Andrew R Bottrill
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, UK
| | - Anthony A Holder
- Malaria Parasitology Laboratory, Francis Crick Institute, London, UK
| | - David S Guttery
- School of Life Sciences, University of Nottingham, Nottingham, UK
- Department of Genetics, Genomics and Cancer Sciences, University of Leicester, Leicester, UK
| | - Rita Tewari
- School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
233
|
Folahan JT, Barabutis N. NEK kinases in cell cycle regulation, DNA damage response, and cancer progression. Tissue Cell 2025; 94:102811. [PMID: 40037068 PMCID: PMC11912005 DOI: 10.1016/j.tice.2025.102811] [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: 11/20/2024] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
The NIMA-related kinase (NEK) family of serine/threonine kinases is essential for the regulation of cell cycle progression, mitotic spindle assembly, and genomic stability. In this review, we explore the structural and functional diversity of NEK kinases, highlighting their roles in both canonical and non-canonical cellular processes. We examine recent preclinical findings on NEK inhibition, showcasing promising results for NEK-targeted therapies, particularly in cancer types characterized by high NEK expression. We discussed the therapeutic potential of targeting NEKs as modulators of cell cycle and DDR pathways, with a focus on identifying strategies to exploit NEK activity for enhanced treatment efficacy. Future research directions are proposed to further elucidate NEK-mediated mechanisms and to develop selective inhibitors that target NEK-related pathways.
Collapse
Affiliation(s)
- Joy T Folahan
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
234
|
Tulin G, Méndez AA, Figueroa NR, Smith C, Folmer MP, Serra D, Wade JT, Checa SK, Soncini FC. Integration of BrfS into the biofilm-controlling cascade promotes sessile Salmonella growth at low temperatures. Biofilm 2025; 9:100254. [PMID: 39927094 PMCID: PMC11804604 DOI: 10.1016/j.bioflm.2025.100254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 02/11/2025] Open
Abstract
Biofilm formation is stimulated by different stress-related physiological and environmental conditions. In Salmonella and Escherichia coli, curli fibers and phosphoethanolamine-cellulose are the major extracellular components of biofilms. The production of both is under the control of CsgD, a transcriptional regulator whose expression is modulated by a number of factors responding to different signals. The atypical MerR-like regulator MlrA is key in the activation of csgD transcription in both Salmonella and E. coli. Recently, MlrB, a SPI-2-encoded MlrA-like regulator that counteracts MlrA by repressing csgD transcription and biofilm formation inside macrophages was identified. Here, we characterize STM1266, a Salmonella-specific MlrA-like regulator, recently renamed BrfS. In contrast to mlrA, brfS transcription increases in minimal growth media and at 20 °C, a temperature not commonly tested in laboratories. Under these conditions, as well as in salt-limited rich medium, deletion or overexpression of brfS affects extracellular matrix production. Using transcriptomics, we uncovered genes under BrfS control relevant for biofilm formation such as csgB and bapA. Transcriptional analysis of these genes in mutants lacking brfS, csgD or both, indicates that BrfS controls curli biosynthesis both in a CsgD-dependent and independent manner. By contrast, at low temperatures, bapA transcription depends only on BrfS, and neither deletion of csgD nor of mlrA modify its expression. Based on these results, we propose that BrfS contributes to Salmonella persistence in the environment, where the pathogen encounters low temperatures and nutrient limitation.
Collapse
Affiliation(s)
- Gonzalo Tulin
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Andrea A.E. Méndez
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Nicolás R. Figueroa
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
- Current position: Centro de Estudios Fotosintéticos y Bioquímicos, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Carol Smith
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - María P. Folmer
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Diego Serra
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Joseph T. Wade
- Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
- RNA Institute, University at Albany, SUNY, Albany, NY, USA
| | - Susana K. Checa
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| | - Fernando C. Soncini
- Instituto de Biología Molecular y Celular de Rosario, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas, Rosario, Argentina
| |
Collapse
|
235
|
Lin LW, Jang HS, Song Z, Ebrahimi A, Yang J, Nguyen BD, O'Donnell EF, Hendrix DA, Maier CS, Kolluri SK. Suppression of global protein synthesis and hepatocellular carcinoma cell growth by Benzimidazoisoquinoline, 4,11-Dichloro-BBQ. Biochem Pharmacol 2025; 236:116896. [PMID: 40157458 DOI: 10.1016/j.bcp.2025.116896] [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: 11/27/2024] [Revised: 02/15/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor best known for mediating biological responses to a wide range of xenobiotics, such as dioxins and polycyclic aromatic hydrocarbons. Recently, AhR has emerged as an important player in cancer biology, with the potential for therapeutic applications through targeted modulation of its activity in specific cancer types. In this study, we report that 4,11-dichloro-BBQ (DiCl-BBQ), a benzimidazoisoquinoline, exhibits AhR-mediated antiproliferative activity in HepG2 hepatocellular carcinoma cells. DiCl-BBQ was found to decrease cell growth at nanomolar concentrations, and this antiproliferative effect persisted even after the compound's removal. Using inducible shRNA expression system, we demonstrated that the inhibitory effect of DiCl-BBQ was significantly reduced following AhR knockdown. Flow cytometric analysis revealed that DiCl-BBQ halted cell division and induced G1 cell cycle arrest in an AhR-dependent manner. Proteomic profiling identified the top four enriched pathways following DiCl-BBQ exposure: metabolism of RNA, translation, ribonucleoprotein complex biogenesis, and carboxylic acid metabolic processes. Notably, DiCl-BBQ caused a dramatic downregulation of translation-associated proteins, with this response diminished in AhR-depleted cells. Consistently, global protein synthesis was significantly repressed in DiCl-BBQ-treated cells. Together, these results indicate that DiCl-BBQ effectively inhibits HepG2 cells growth by inducing G1 cell cycle arrest and downregulating the protein translation machinery in an AhR-dependent manner.
Collapse
Affiliation(s)
- Lo-Wei Lin
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Hyo Sang Jang
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Zifeng Song
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Arpa Ebrahimi
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Jun Yang
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA
| | - Bach D Nguyen
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Edmond F O'Donnell
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - David A Hendrix
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331, USA; School of Electrical Engineering and Computer Science, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Siva K Kolluri
- Cancer Research Laboratory, Department of Environmental & Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
236
|
Zhang J, Guo H, Gong C, Shen J, Jiang G, Liu J, Liang T, Guo L. Therapeutic targets in the Wnt signaling pathway: Treating cancer with specificity. Biochem Pharmacol 2025; 236:116848. [PMID: 40049295 DOI: 10.1016/j.bcp.2025.116848] [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: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
The Wnt signaling pathway is a critical regulatory mechanism that governs cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, and the tumor immune microenvironment, while also maintaining tissue homeostasis. Dysregulated activation of this pathway is implicated in various cancers, closely linked to tumor initiation, progression, and metastasis. The Wnt/β-catenin axis plays a central role in the pathogenesis of common cancers, including colorectal cancer (CRC), breast cancer (BC), liver cancer, and lung cancer. Unlike traditional chemotherapy, targeted therapy offers a more precise approach to cancer treatment. As a key regulator of oncogenesis, the Wnt pathway represents a promising target for clinical interventions. This review provides a comprehensive analysis of the Wnt signaling pathway, exploring its roles in tumor biology and its implications in human malignancies. It further examines the molecular mechanisms and modes of action across different cancers, detailing how the Wnt pathway contributes to tumor progression through mechanisms such as metastasis promotion, immune modulation, drug resistance, and enhanced cellular proliferation. Finally, therapeutic strategies targeting Wnt pathway components are discussed, including inhibitors targeting extracellular members, as well as those within the cell membrane, cytoplasm, and nucleus. The potential of these targets in the development of novel therapeutic agents underscores the critical importance of intervening in the Wnt signaling pathway for effective cancer treatment.
Collapse
Affiliation(s)
- Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Chengxuan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jie Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guijie Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiarui Liu
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China.
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
237
|
Madhukar G, Haque MA, Khan S, Kim JJ, Danishuddin. E3 ubiquitin ligases and their therapeutic potential in disease Management. Biochem Pharmacol 2025; 236:116875. [PMID: 40120724 DOI: 10.1016/j.bcp.2025.116875] [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: 11/27/2024] [Revised: 02/05/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Ubiquitination is a vital post-translational modification that regulates protein stability and various cellular processes through the addition of ubiquitin molecules. Central to this process are E3 ubiquitin ligases, which determine the specificity of ubiquitination by coordinating the attachment of ubiquitin to target proteins, influencing their degradation, localization, and activity. E3 ubiquitin ligases are involved in numerous cellular pathways, including DNA repair, cell proliferation, and immune responses. Dysregulation of E3 ubiquitin ligases is often associated with cancer, contributing to tumor progression and resistance to therapies. The development of targeted protein degraders, such as proteolysis-targeting chimeras (PROTACs), represents a significant advancement in drug discovery, leveraging the specificity of E3 ubiquitin ligases to selectively eliminate pathogenic proteins. However, challenges remain in translating this knowledge into effective therapies, including issues related to tissue-specific targeting and off-target effects. The limitations also include a limited understanding of ligase-substrate interactions that includes both the identification of novel E3 ligases and their substrates, as well as understanding the dynamic, context-dependent nature of these interactions, which can vary across tissue types or disease states This review emphasizes the therapeutic potential of E3 ubiquitin ligases, exploring their diverse roles in disease, their contribution to targeted degradation strategies while highlighting the need for further research to overcome current limitations and enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Geet Madhukar
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, USA
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Shawez Khan
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| | - Danishuddin
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
238
|
Cui J, Shen Y, Song Z, Fan D, Hu B. Mechanism by which Rab5 promotes regeneration and functional recovery of zebrafish Mauthner axons. Neural Regen Res 2025; 20:1816-1824. [PMID: 39104118 PMCID: PMC11688562 DOI: 10.4103/nrr.nrr-d-23-00529] [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: 04/04/2023] [Revised: 08/02/2023] [Accepted: 01/26/2024] [Indexed: 08/07/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202506000-00031/figure1/v/2024-08-05T133530Z/r/image-tiff Rab5 is a GTPase protein that is involved in intracellular membrane trafficking. It functions by binding to various effector proteins and regulating cellular responses, including the formation of transport vesicles and their fusion with the cellular membrane. Rab5 has been reported to play an important role in the development of the zebrafish embryo; however, its role in axonal regeneration in the central nervous system remains unclear. In this study, we established a zebrafish Mauthner cell model of axonal injury using single-cell electroporation and two-photon axotomy techniques. We found that overexpression of Rab5 in single Mauthner cells promoted marked axonal regeneration and increased the number of intra-axonal transport vesicles. In contrast, treatment of zebrafish larvae with the Rab kinase inhibitor CID-1067700 markedly inhibited axonal regeneration in Mauthner cells. We also found that Rab5 activated phosphatidylinositol 3-kinase (PI3K) during axonal repair of Mauthner cells and promoted the recovery of zebrafish locomotor function. Additionally, rapamycin, an inhibitor of the mechanistic target of rapamycin downstream of PI3K, markedly hindered axonal regeneration. These findings suggest that Rab5 promotes the axonal regeneration of injured zebrafish Mauthner cells by activating the PI3K signaling pathway.
Collapse
Affiliation(s)
- Jiantao Cui
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Yueru Shen
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Zheng Song
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Dinggang Fan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Bing Hu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
239
|
Pandey A, Goswami A, Jithin B, Shukla S. Autophagy: The convergence point of aging and cancer. Biochem Biophys Rep 2025; 42:101986. [PMID: 40224538 PMCID: PMC11986642 DOI: 10.1016/j.bbrep.2025.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy, a dynamic intracellular degradation system, is critical for cellular renovation and maintaining equilibrium. By eliminating damaged components and recycling essential molecules, autophagy safeguards cellular integrity and function. The versatility of the autophagy process across various biological functions enable cells to adapt and maintain homeostasis under unfavourable conditions. Disruptions in autophagy can shift a cell from a healthy state to a disease state or, conversely, support a return to health. This review delves into the multifaceted role of autophagy during aging and age-related diseases such as cancer, highlighting its significance as a unifying target with promising therapeutic implications. Cancer development is a dynamic process characterized by the acquisition of diverse survival capabilities for proliferating at different stages. This progression unfolds over time, with cancer cells exploiting autophagy to overcome encountered stress conditions during tumor development. Notably, there are several common pathways that utilize the autophagy process during aging and cancer development. This highlights the importance of autophagy as a crucial therapeutic target, holding the potential to not only impede the growth of tumor but also enhance the patient's longevity. This review aims to simplify the intricate relationship between cancer and aging, with a particular focus on the role of autophagy.
Collapse
Affiliation(s)
- Anchala Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| | | | | | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh, 462066, India
| |
Collapse
|
240
|
Chen S, Li P, Shi K, Tang S, Zhang W, Peng C, Li T, Xie H, Liu C, Zhou J. Tanshinone IIA promotes ferroptosis in cutaneous melanoma via STAT1-mediated upregulation of PTGS2 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156702. [PMID: 40222167 DOI: 10.1016/j.phymed.2025.156702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Melanoma is highly aggressive, metastatic with a poor prognosis. Despite significant advances in targeted therapies and immunotherapies, their efficiency limited by drug resistance. Tanshinone IIA (Tan IIA), a bioactive compound derived from Traditional Chinese plant, exhibits significant anticancer potential, which still needs more research in its complex regulatory mechanisms. PURPOSE This study aimed to elucidate the putative targets and regulatory mechanisms of Tan IIA in anti-melanoma, with a focus on its role in inducing ferroptosis. STUDY DESIGN We designed the experiment to explore the effects of Tan IIA on melanoma through both in vitro and in vivo experiments and to investigate the underlying mechanisms through transcriptomics combining network pharmacology analysis. METHOD Ferroptosis monitored by Malondialdehyde (MDA), Fe2+, reactive oxygen species (ROS) and glutathione (GSH) in vivo and in vitro. RNA sequence was performed to explore the key regulatory pathways involved in Tan IIA-induced ferroptosis. Chromatin immunoprecipitation (ChIP) and Luciferase assays were used to validate transcription factor responsible for prostaglandin-endoperoxide synthase 2 (PTGS2) regulation. Additionally, RT-qPCR, western blot, IF, IHC were aimed to evaluate the expression of target gene. RESULT Tan IIA markedly suppresses melanoma growth in a xenograft model. The same effect performed on inhibition melanoma cells and promotion to ferroptosis with accumulation of ROS, MDA, and Fe²⁺levels and GSH consumption. RNA sequencing and public database analysis revealed that Tan IIA regulates PTGS2, the critical marker of ferroptosis, and PTGS2-knockdown attenuates Tan IIA -induced ferroptosis in melanoma cells. Furthermore, we identified that Tan IIA stimulate signal transducer and activator of transcription 1 (STAT1), a transcription factor, promoting PTGS2 expression and localized in the cell cytoplasm. Moreover, downregulation of the transcription factor STAT1 lead to PTGS2 downregulation and also inhibit ferroptosis in melanoma. CONCLUSION This study, the first to link Tan IIA-induced ferroptosis to the STAT1/PTGS2 axis in melanoma, identifies STAT1 and PTGS2 as novel therapeutic targets for melanoma, which demonstrates the potential of natural compounds Tan IIA in overcoming drug resistance and integrates traditional medicine with advanced molecular techniques for mechanistic exploration.
Collapse
Affiliation(s)
- Shuyue Chen
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Peiting Li
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Ke Shi
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Cong Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Tianyu Li
- Department of Burns and Plastic Surgery, Nanshi Hospital of Nanyang, Nanyang, Henan Province 473000, China
| | - Huiqing Xie
- Department of Rehabilitation, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Can Liu
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| | - Jianda Zhou
- Department of Plastic and Reconstructive Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China.
| |
Collapse
|
241
|
Yang C, Wu H, Hao R, Liao Y, Wang Q, Deng Y. Integrated transcriptomic and lipidomic analysis reveals that arachidonic acid mediates the allograft-induced stress response in Pinctada fucata martensii. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 54:101411. [PMID: 39765130 DOI: 10.1016/j.cbd.2024.101411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/28/2024] [Accepted: 12/28/2024] [Indexed: 03/12/2025]
Abstract
This study investigated the protective effect of arachidonic acid (ARA) against the allograft-induced stress response in Pinctada fucata martensii by characterizing pearl production traits and changes in genes and lipids during postoperative care. Survival and pearl production traits were higher in the ARA treatment group (ARAG) than in the control group (CG). There were 1536 differentially expressed genes (DEGs) in CG-1d vs ARAG-1d and 833 DEGs in CG-3d vs ARAG-3d. DEGs in CG-1d vs ARAG-1d were mainly enriched in "NOD-like receiver signaling pathway," "Glycerolipid metabolism," and "Sphingolipid metabolism." DEGs in CG-3d vs ARAG-3d were mainly enriched in "Apoptosis" and "ARA metabolism." Lipidomics analysis revealed 36 types of lipids. The PC, LPE (22:5), and LPE (18:0) content at 3 d after implantation was lower in the ARAG than in the CG. The PS (37,1) content was significantly higher at 3 d after implantation and the content of ceramides was significantly lower at 1 and 3 d after implantation in the ARAG than in the CG. The results indicated that ARA may alter the composition of lipids, modify the unsaturation of lipids, and regulate apoptosis, immunity, and lipid metabolism in pearl oysters, which enhances pearl production traits. These findings provide theoretical and practical basis for further alleviating the inflammatory response of pearl oysters after implantation.
Collapse
Affiliation(s)
- Chuangye Yang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Hailing Wu
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ruijuan Hao
- Development and Research Center for Biological Marine Resources, Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524006, China
| | - Yongshan Liao
- Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Center of Guangdong Province, Zhanjiang 524088, China
| | - Qingheng Wang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Center of Guangdong Province, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy culture, Zhanjiang 524088, China
| | - Yuewen Deng
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Guangdong Science and Innovation Center for Pearl Culture, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Center of Guangdong Province, Zhanjiang 524088, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy culture, Zhanjiang 524088, China.
| |
Collapse
|
242
|
Aloliqi AA, Alnuqaydan AM, Albutti A, Alharbi BF, Rahmani AH, Khan AA. Current updates regarding biogenesis, functions and dysregulation of microRNAs in cancer: Innovative approaches for detection using CRISPR/Cas13‑based platforms (Review). Int J Mol Med 2025; 55:90. [PMID: 40242952 PMCID: PMC12021393 DOI: 10.3892/ijmm.2025.5531] [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: 01/15/2025] [Accepted: 03/04/2025] [Indexed: 04/18/2025] Open
Abstract
MicroRNAs (miRNAs) are short non‑coding RNAs, which perform a key role in cellular differentiation and development. Most human diseases, particularly cancer, are linked to miRNA functional dysregulation implicated in the expression of tumor‑suppressive or oncogenic targets. Cancer hallmarks such as continued proliferative signaling, dodging growth suppressors, invasion and metastasis, triggering angiogenesis, and avoiding cell death have all been demonstrated to be affected by dysregulated miRNAs. Thus, for the treatment of different cancer types, the detection and quantification of this type of RNA is significant. The classical and current methods of RNA detection, including northern blotting, reverse transcription‑quantitative PCR, rolling circle amplification and next‑generation sequencing, may be effective but differ in efficiency and accuracy. Furthermore, these approaches are expensive, and require special instrumentation and expertise. Thus, researchers are constantly looking for more innovative approaches for miRNA detection, which can be advantageous in all aspects. In this regard, an RNA manipulation tool known as the CRISPR and CRISPR‑associated sequence 13 (CRISPR/Cas13) system has been found to be more advantageous in miRNA detection. The Cas13‑based miRNA detection approach is cost effective and requires no special instrumentation or expertise. However, more research and validation are required to confirm the growing body of CRISPR/Cas13‑based research that has identified miRNAs as possible cancer biomarkers for diagnosis and prognosis, and as targets for treatment. In the present review, current updates regarding miRNA biogenesis, structural and functional aspects, and miRNA dysregulation during cancer are described. In addition, novel approaches using the CRISPR/Cas13 system as a next‑generation tool for miRNA detection are discussed. Furthermore, challenges and prospects of CRISPR/Cas13‑based miRNA detection approaches are described.
Collapse
Affiliation(s)
- Abdulaziz A. Aloliqi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Abdullah M. Alnuqaydan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Aqel Albutti
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Basmah F. Alharbi
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia
| |
Collapse
|
243
|
Amarsanaa E, Wie M, Shin U, Bilal N, Hwang J, Lee E, Lee S, Kim BG, Kim S, Lee Y, Myung K. Synergistic enhancement of PARP inhibition via small molecule UNI66-mediated suppression of BRD4-dependent transcription of RAD51 and CtIP. NAR Cancer 2025; 7:zcaf013. [PMID: 40308947 PMCID: PMC12041917 DOI: 10.1093/narcan/zcaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 02/18/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Targeted therapy leveraging synthetic lethality in homologous recombination (HR)-defective tumors, particularly in BRCA-mutated tumors through poly(ADP-ribose) polymerase (PARP)-dependent repair inhibition, has shown success. However, the challenge lies in the ability of the tumors to reactivate HR via diverse mechanisms, leading to resistance against PARP-dependent repair inhibition. Addressing this issue, the down-regulation of HR activity has been explored as a potential strategy to overcome PARP inhibitor-resistant tumors. Yet, the intricate modulation of HR gene expression in mammalian cells is still not fully understood. In this study, we used a small molecule, UNI66, identified from high-throughput screening, to investigate regulatory mechanisms of HR. UNI66 was observed to induce synthetic lethality in PARP1-deficient cells and enhanced the sensitivity of multiple cancer cells to PARP inhibitors, suggesting a role in HR down-regulation. Mechanistically, UNI66 was found to interact with and inhibit BRD4 protein binding to the promoters of CtIP and RAD51 genes, resulting in the down-regulation of their transcription. This decrease in CtIP and RAD51 expression was associated with reduced HR activity, thereby increasing the sensitivity of tumors to PARP inhibitors. These findings indicate that BRD4-mediated transcriptional regulation of CtIP and RAD51 influences HR activity, which may have implications for overcoming resistance to PARP inhibitors.
Collapse
Affiliation(s)
- Enkhzul Amarsanaa
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Minwoo Wie
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Unbeom Shin
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Nabeela Bilal
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jungme Hwang
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Eun A Lee
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Seon Young Lee
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Byung-Gyu Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Shinseog Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Yoonsung Lee
- Clinical Research Institute, Kyung Hee University Hospital at Gangdong, School of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| |
Collapse
|
244
|
Thakur V, Islam MM, Singh S, Rathore S, Muneer A, Dutta G, Banday MM, Arora P, Hossain ME, Jain S, Ali S, Mohmmed A. A dynamin-like protein in Plasmodium falciparum plays an essential role in parasite growth, mitochondrial development and homeostasis during asexual blood stages. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119940. [PMID: 40157510 DOI: 10.1016/j.bbamcr.2025.119940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Malaria parasites harbour a single mitochondrion, and its proper segregation during parasite multiplication is crucial for the propagation of the parasite within the host. Mitochondrial division machinery consists of several proteins that associate with the mitochondrial membrane during segregation. Here, we have identified a dynamin-like protein in P. falciparum, PfDyn2, and deciphered its role in mitochondrial growth and homeostasis. A GFP targeting approach combined with high-resolution microscopy studies showed that the PfDyn2 associates with the mitochondrial membrane at specific sites during mitochondrial division. The C-terminal degradation tag mediated inducible knock-down (iKD) of PfDyn2 significantly inhibited parasite growth. PfDyn2-iKD hindered mitochondrial development and functioning, decreased mtDNA replication, and induced mitochondrial oxidative stress, ultimately causing parasite death. Regulated overexpression of a phosphorylation mutant of PfDyn2 (Ser-612-Ala) did not affect the recruitment of PfDyn2 on the mitochondria; normal mitochondrial division and parasite growth showed that phosphorylation/dephosphorylation of this conserved serine residue (Ser612) may not be responsible for regulating recruitment of PfDyn2 to the mitochondrion. Overall, we show the essential role of PfDyn2 in mitochondrial development and maintaining its homeostasis during the asexual cycle of the parasite.
Collapse
Affiliation(s)
- Vandana Thakur
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Md Muzahidul Islam
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Shweta Singh
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sumit Rathore
- All India Institute of Medical Sciences, New Delhi 110029, India
| | - Azhar Muneer
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Gaurav Dutta
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Mudassir M Banday
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Priya Arora
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Mohammad E Hossain
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Shaifali Jain
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India.
| |
Collapse
|
245
|
Lin R, Zhang W, Tian R, Zhang L, Hong J, Wang L, Kang H, Yu J, Zhou Y. CPK27 enhances cold tolerance by promoting flavonoid biosynthesis through phosphorylating HY5 in tomato. THE NEW PHYTOLOGIST 2025; 246:2174-2191. [PMID: 40235338 DOI: 10.1111/nph.70134] [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: 10/26/2024] [Accepted: 03/23/2025] [Indexed: 04/17/2025]
Abstract
Cold stress is a major environmental challenge affecting the production of crops. Calcium-dependent protein kinases (CDPKs/CPKs) are crucial regulators relaying calcium (Ca2+) signals into cellular stress responses. However, the specific mechanisms of CPKs in regulating cold stress signaling are not well understood. In this study, through genetic, physiological and molecular biology assays, we characterized the function of CPK27 in enhancing tomato cold tolerance. We found that CPK27 stimulates flavonoid biosynthesis in a Ca2+-dependent manner, which in turn boosts the plant's tolerance. Tomato plants lacking CPK27 (cpk27) showed decreased flavonoid levels under cold stress, accompanied by the increased sensitivity to cold. Activated by cold stress, CPK27 accumulates within the nucleus, where it physically interacts and phosphorylates ELONGATED HYPOCOTYL 5 (HY5) protein at serine23 (S23) and S57 residues, contributing to the cold-induced accumulation of HY5 protein. HY5 directly binds to the promoter regions and stimulates the transcription of flavonoid biosynthesis genes. Further genetic analysis showed that CPK27 acts upstream of HY5, and the flavonoid biosynthesis pathway activated by CPK27 is HY5-dependent. Our study elucidates the regulatory mechanism whereby the CPK27-HY5 molecule integrates cold-triggered Ca2+ signals with flavonoid biosynthesis pathways to confer cold stress tolerance, thereby uncovering the key strategy for cold signal transduction.
Collapse
Affiliation(s)
- Rui Lin
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Wenjing Zhang
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Rong Tian
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Limeng Zhang
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Jiachen Hong
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Lingyu Wang
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
- Agricultural Experiment Station, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Huijia Kang
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Jingquan Yu
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
- Hainan Institute, Zhejiang University, Sanya, 572025, China
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Yanhong Zhou
- Department of Horticulture, Zhejiang University, Zijingang Campus, 866 Yuhangtang Road, Hangzhou, 310058, China
- Hainan Institute, Zhejiang University, Sanya, 572025, China
- Zhejiang Key Laboratory of Horticultural Crop Quality Improvement, 866 Yuhangtang Road, Hangzhou, 310058, China
| |
Collapse
|
246
|
Fujita Y, De Velasco MA, Hayashi H, Nakagawa K, Nishio K. Exploration of genes related to the development of cancer of unknown primary. Oncol Rep 2025; 53:72. [PMID: 40314076 DOI: 10.3892/or.2025.8905] [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: 11/19/2024] [Accepted: 02/21/2025] [Indexed: 05/03/2025] Open
Abstract
The biological basis of the development of cancer of unknown primary (CUP) remains largely unknown, with no evidence of whether a common biological basis exists at present. Our previous multicenter clinical study predicted the primary site of CUP for site‑specific therapy. Concomitantly with the study, a microarray analysis of tumor mRNA samples obtained from 60 participants of the study with CUP was performed, and a gene expression profile specific to CUP was constructed. Several of the genes identified as being upregulated/downregulated in CUP could potentially be clinically useful common biomarkers of CUP. In the present study, to identify genes that may be more closely related to the development of CUP (characterized by its metastatic potential) among the upregulated genes, cell‑based small interfering RNA screening was performed in vitro, and two genes, protein kinase DNA‑activated catalytic subunit (PRKDC) and proteasome subunit β type‑4 (PSMB4), were identified to be possibly involved in the metastatic ability of CUP, since knockdown of these genes resulted in reduced migration of A549 cells. These genes were further knocked down in A549 cells using short hairpin RNAs (shRNAs) and the cells were implanted into the footpad of mice. Marked suppression of the metastatic ability of implanted cells from the footpad to the popliteal lymph node (LN) was observed in cells transfected with the shRNAs for PRKDC and PSMB4. In addition, bortezomib, a proteasome inhibitor, markedly reduced the ability of cells implanted into the footpad to metastasize to the LNs, as well as cell growth at the metastatic site, compared with vehicle or NU7447 (inhibitor of PRKDC). These findings indicated that proteasomal function activation augmented the metastatic ability of malignant CUP cells.
Collapse
Affiliation(s)
- Yoshihiko Fujita
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka‑Sayama, Osaka 589‑8511, Japan
| | - Marco A De Velasco
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka‑Sayama, Osaka 589‑8511, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka‑Sayama, Osaka 589‑8511, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka‑Sayama, Osaka 589‑8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka‑Sayama, Osaka 589‑8511, Japan
| |
Collapse
|
247
|
Fullstone T, Rohm H, Kaltofen T, Hierlmayer S, Reichenbach J, Schweikert S, Knodel F, Loeffler AK, Mayr D, Jeschke U, Mahner S, Kessler M, Trillsch F, Rathert P. Identification of FLYWCH1 as a regulator of platinum-resistance in epithelial ovarian cancer. NAR Cancer 2025; 7:zcaf012. [PMID: 40191655 PMCID: PMC11970373 DOI: 10.1093/narcan/zcaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/09/2025] Open
Abstract
Platinum-based combination chemotherapy remains the backbone of first-line treatment for patients with advanced epithelial ovarian cancer (EOC). While most patients initially respond well to the treatment, patients with relapse ultimately develop platinum resistance. This study identified FLYWCH-type zinc finger-containing protein 1 (FLYWCH1) as an important regulator in the resistance development process. We showed that the loss of FLYWCH1 promotes platinum resistance in EOC cells, and the low FLYWCH1 expression is correlated with poor prognosis of EOC patients. In platinum-sensitive cells, FLYWCH1 colocalizes with H3K9me3, but this association is significantly reduced when cells acquire resistance. The suppression of FLYWCH1 induces gene expression changes resulting in the deregulation of pathways associated with resistance. In line with its connection to H3K9me3, FLYWCH1 induces gene silencing in a synthetic reporter assay and the suppression of FLYWCH1 alters H3K9me3 at promoter regions and repeat elements. The loss of FLYWCH1 leads to the derepression of LTR and Alu repeats, thereby increasing transcriptional plasticity and driving the resistance development process. Our data highlight the importance of FLYWCH1 in chromatin biology and acquisition of platinum resistance through transcriptional plasticity and propose FLYWCH1 as a potential biomarker for predicting treatment responses in EOC patients.
Collapse
MESH Headings
- Female
- Humans
- Drug Resistance, Neoplasm/genetics
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/drug therapy
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Cell Line, Tumor
- Histones/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Drosophila Proteins/genetics
- Drosophila Proteins/metabolism
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/pathology
- Neoplasms, Glandular and Epithelial/metabolism
- Platinum/pharmacology
- Prognosis
- Promoter Regions, Genetic
- Antineoplastic Agents/pharmacology
Collapse
Affiliation(s)
- Tabea L Fullstone
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Helene Rohm
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Till Kaltofen
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sophia Hierlmayer
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Juliane Reichenbach
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Simon Schweikert
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Franziska Knodel
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Ann-Kathrin Loeffler
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| | - Doris Mayr
- Institute of Pathology, LMU Munich, 81377 Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
- Department of Obstetrics and Gynaecology, University Hospital Augsburg, 86156 Augsburg, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Mirjana Kessler
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynaecology, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Philipp Rathert
- Department of Molecular Biochemistry, Institute of Biochemistry, University of Stuttgart, 70569 Stuttgart, Germany
| |
Collapse
|
248
|
Cros-Perrial E, Beaumel S, Gimbert M, Camus N, Vicente C, Sekiou I, Figuet L, Duruisseaux M, Dumontet C, Jordheim LP. SLX4 and XPF are involved in cell migration and EMT in a cell-specific manner. Biochem Pharmacol 2025; 236:116885. [PMID: 40118290 DOI: 10.1016/j.bcp.2025.116885] [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: 11/20/2024] [Revised: 02/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
SLX4 and XPF are two proteins involved in DNA repair, but very little is known about their potential roles in other processes of cancer cell biology. We developed original cell models with CRISPR-Cas9-mediated knock-out of SLX4 and/or XPF using five different cell lines (A549, NCI-H1703, COLO-357, HT-29 and HEK-293 T), and performed characterization with cell biology experiments including migration assays, drug sensitivity testing, cell proliferation assessment and Western blots for relevant proteins. Results showed decreased migration of all models in HT-29 cells, of XPF-deficient COLO-357 cells and of SLX4-deficient HEK-293 T cells. Modified cell models had overall increased sensitivity to cisplatin and mitomycine C, and some models showed an increased frequency of double-stranded DNA damages. One NCI-H1703 cell model showed major karyotypic modifications, and epithelial to mesenchymal transition (EMT)-related proteins were modified in several models. Finally, knocking out one or both proteins in A549 cells had not the same impact on in vivo growth in mice. These original cell models allowed us to identify new and DNA repair-unrelated cellular roles of SLX4 and XPF in cancer cell biology. Our results should be considered within work on Nucleotide Excision Repair (NER) inhibition targeting SLX, XPF or other related proteins.
Collapse
Affiliation(s)
- Emeline Cros-Perrial
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Sabine Beaumel
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Manon Gimbert
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Ninon Camus
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Clara Vicente
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Imane Sekiou
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Léa Figuet
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France
| | - Michaël Duruisseaux
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Charles Dumontet
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France; Hospices Civils de Lyon, Lyon, France
| | - Lars Petter Jordheim
- Université Claude Bernard Lyon 1, INSERM U-1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon 69008 Lyon, France.
| |
Collapse
|
249
|
Lin W, Li Y, Huang H, Zhao P, Su Y, Fang CY. Harmine hydrochloride induces G0/G1 cell cycle arrest and apoptosis in oral squamous carcinoma cells. Exp Ther Med 2025; 29:111. [PMID: 40242602 PMCID: PMC12001316 DOI: 10.3892/etm.2025.12861] [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: 12/06/2024] [Accepted: 02/26/2025] [Indexed: 04/18/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents the most frequently occurring form of oral cancer. However, despite the availability of advanced treatment modalities, the global 5-year survival rate for patients with advanced OSCC remains at ~50-60%. Devising alternative therapeutic strategies for oral cancer has therefore become an urgent need. Harmine, a β-carboline alkaloid, has recently been shown to exhibit anticancer activity. Compared with harmine, harmine hydrochloride (HH), a derivative of harmine, has improved water solubility and stability, so can absorb into tissues more readily. Therefore, the present study aimed to investigate the anticancer activity of HH in OSCC cells. A Cell Counting Kit-8 assay was performed to assess the cytotoxic effects of HH on the OSCC cell lines, SCC-4 and SCC-25. Flow cytometric analysis was subsequently employed to examine both the cell cycle profile and the extent of apoptosis. Western blotting was used to assess the expression levels of the regulatory proteins involved in these biological activities, and treatment with a pan-caspase inhibitor (Z-VAD-FMK) confirmed the involvement of the apoptotic pathway. Furthermore, western blotting was used to investigate which signaling pathways were affected in the HH-treated cells. Taken together, the findings of the present study demonstrated that HH was cytotoxic in OSCC cells. HH treatment induced G0/G1 phase cell cycle arrest and apoptosis. Additionally, the MAPK pathway was shown to be involved in HH-induced apoptosis in SCC-4 cells. Therefore, HH exhibited anticancer activity, and may be a putative therapeutic agent for the treatment of OSCC in the future.
Collapse
Affiliation(s)
- Weiting Lin
- Department of Stomatology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Yizhen Li
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Hsinyi Huang
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Peiwen Zhao
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Yining Su
- Department of Stomatology, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Chiung-Yao Fang
- Department of Medical Research, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
- Institute of Molecular Biology, National Chung Cheng University, Chiayi 621, Taiwan, R.O.C
| |
Collapse
|
250
|
Yen CC, Chen PCH, Chen SC, Wu WC, Yen CH, Lin YC, Wu PK, Chen CM, Wang JY, Chao TC, Yang MH, Fletcher JA. Ferroptosis as a therapeutic vulnerability in MDM2 inhibition in dedifferentiated liposarcoma. Oncol Lett 2025; 29:269. [PMID: 40247991 PMCID: PMC12005077 DOI: 10.3892/ol.2025.15015] [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: 10/24/2024] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Ferroptosis is a form of necrotic cell death characterized by phospholipid oxidation. The cystine-glutamate antiporter (xCT), composed of solute carrier family 7 member 11 (SLC7A11) and SLC3A2, imports cystine for glutathione synthesis. Glutathione peroxidase 4 (GPX4) requires glutathione to counteract lipid peroxidation and prevent ferroptosis. Erastin, an xCT inhibitor, and Ras-selective lethal small molecule 3 (RSL3), a GPX4 inhibitor, suppress GPX4 function and induce ferroptosis. Tumor protein p53 (TP53) has a paradoxical role in ferroptosis regulation. Mouse double minute 2 homolog (MDM2), a negative regulator of TP53, is a key oncogene in well-differentiated liposarcoma (WDLPS) and dedifferentiated liposarcoma (DDLPS). Therefore, the present study explored the role of ferroptosis in DDLPS treatment response and resistance. Publicly available expression profiles of WDLPS, DDLPS and adipose tissue were analyzed, and the differential expression of ferroptosis-related genes regulated by the MDM2-TP53 pathway was identified in WDLPS and DDLPS. In vitro experiments were performed to assess the effects of erastin and RSL3 on the viability, lipid peroxidation and apoptosis of DDLPS cell lines. The results revealed that erastin and RSL3 induced lipid peroxidation and apoptosis, thereby exerting cytotoxic effects. In addition, nutlin-3, an MDM2 inhibitor, was demonstrated to increase lipid peroxidation and cytotoxicity when applied prior to erastin treatment. Notably, nutlin-3 also upregulated SLC3A2 expression in DDLPS cell lines, thereby enhancing cystine uptake. This increase in cystine uptake was suppressed by erastin. In addition, nutlin-3-induced SLC3A2 upregulation was abolished by TP53 knockdown. Nutlin-3 combined with erastin or RSL3 reduced absolute p-4EBP-1 levels in NDDLS-1 cells and p-p70S6 levels in both cell lines, with no significant impact on the p-4EBP-1/4EBP-1 and p-p70S6/p70S6 ratios. These results indicate that ferroptosis is a therapeutic vulnerability in the response to MDM2 inhibition in DDLPS. Furthermore, combining MDM2 inhibitors with ferroptosis-inducing agents may provide a potential therapeutic strategy for DDLPS and the role of mTOR in the pro-apoptotic effect of these combinations deserve further investigation.
Collapse
Affiliation(s)
- Chueh-Chuan Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Paul Chih-Hsueh Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - San-Chi Chen
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Wen-Chi Wu
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chiao-Han Yen
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Yung-Chan Lin
- Department of Medical Research, Division of Clinical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Po-Kuei Wu
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Chao-Ming Chen
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
| | - Jir-You Wang
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Institute of Traditional Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Ta-Chung Chao
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- Department of Orthopedics and Traumatology, Therapeutical and Research Center of Musculoskeletal Tumor, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Muh-Hwa Yang
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei 112201, Taiwan, R.O.C
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
- Institute of Clinical Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | | |
Collapse
|