51
|
Khan MZ, Zugaza JL, Torres Aleman I. The signaling landscape of insulin-like growth factor 1. J Biol Chem 2025; 301:108047. [PMID: 39638246 PMCID: PMC11748690 DOI: 10.1016/j.jbc.2024.108047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The sheer amplitude of biological actions of insulin-like growth factor I (IGF-1) affecting all types of cells in all tissues suggests a vast signaling landscape for this ubiquitous humoral signal. While the canonical signaling pathways primarily involve the Ras/MAPK and PI3K/AKT cascades, the evolutionary conservation of insulin-like peptides (ILPs) and their pathways hints at the potential for novel functions to emerge over time. Indeed, the evolutionary trajectory of ILPs opens the possibility of either novel functions for these two pathways, novel downstream routes, or both. Evidence supporting this notion includes observations of neofunctionalization in bony fishes or crustaceans, and the involvement of ILPs pathways in invertebrate eusociality or in vertebrate bone physiology, respectively. Such evolutionary processes likely contribute to the rich diversity of ILPs signaling observed today. Moreover, the interplay between conserved signaling pathways, such as those implicated in aging (predominantly involving the PI3K-AKT route), and lesser known pathways, such as those mediated by biased G-protein coupled receptors and others even less known, may underpin the context-dependent actions characteristic of ILPs signaling. While canonical IGF-1 signaling is often assumed to account for the intracellular pathways utilized by this growth factor, a comprehensive analysis of all the pathways mediated by the IGF-1 receptor (IGF-1R) remains lacking. This review aims to explore both canonical and non-canonical routes of IGF-1R action across various cell types, offering a detailed examination of the mechanisms underlying IGF-1 signaling and highlighting the significant gaps in our current understanding.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain
| | - Jose Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain; Ikerbasque Science Foundation, Bilbao, Spain
| | - Ignacio Torres Aleman
- Achucarro Basque Center for Neuroscience, Leioa, Spain; CIBERNED, Madrid, Spain; Ikerbasque Science Foundation, Bilbao, Spain.
| |
Collapse
|
52
|
Li X, Wu C, Lu X, Wang L. Predictive models of sarcopenia based on inflammation and pyroptosis-related genes. Front Genet 2024; 15:1491577. [PMID: 39777262 PMCID: PMC11703911 DOI: 10.3389/fgene.2024.1491577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Background Sarcopenia is a prevalent condition associated with aging. Inflammation and pyroptosis significantly contribute to sarcopenia. Methods Two sarcopenia-related datasets (GSE111016 and GSE167186) were obtained from the Gene Expression Omnibus (GEO), followed by batch effect removal post-merger. The "limma" R package was utilized to identify differentially expressed genes (DEGs). Subsequently, LASSO analysis was conducted on inflammation and pyroptosis-related genes (IPRGs), resulting in the identification of six hub IPRGs. A novel skeletal muscle aging model was developed and validated using an independent dataset. Additionally, Gene Ontology (GO) enrichment analysis was performed on DEGs, along with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and gene set enrichment analysis (GSEA). ssGSEA was employed to assess differences in immune cell proportions between healthy muscle groups in older versus younger adults. The expression levels of the six core IPRGs were quantified via qRT-PCR. Results A total of 44 elderly samples and 68 young healthy samples were analyzed for DEGs. Compared to young healthy muscle tissue, T cell infiltration levels in aged muscle tissue were significantly reduced, while mast cell and monocyte infiltration levels were relatively elevated. A new diagnostic screening model for sarcopenia based on the six IPRGs demonstrated high predictive efficiency (AUC = 0.871). qRT-PCR results indicated that the expression trends of these six IPRGs aligned with those observed in the database. Conclusion Six biomarkers-BTG2, FOXO3, AQP9, GPC3, CYCS, and SCN1B-were identified alongside a diagnostic model that offers a novel approach for early diagnosis of sarcopenia.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Wu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang Lu
- Department of Geriatrics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Wang
- Department of Geriatrics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
53
|
Feng M, Gao B, Garcia LR, Sun Q. Bacterial purine metabolism modulates C. elegans development and stress tolerance via DAF-16. FEBS J 2024. [PMID: 39708289 DOI: 10.1111/febs.17363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
The purine metabolism is crucial for cellular function and is a conserved metabolic network from prokaryotes to humans. While extensively studied in microorganisms like yeast and bacteria, the impact of perturbing dietary intermediates from the purine biosynthesis on animal development and growth remains poorly understood. We utilized Caenorhabditis elegans as the metazoan model to investigate the mechanisms underlying this deficiency. Through a high-throughput screening of an Escherichia coli mutant library Keio collection, we identified 34 E. coli mutants that delay C. elegans development. Among these mutants, we found that E. coli purE gene is an essential genetic component that promotes host development in a dose-dependent manner. Further metabolites supplementation suggests that bacterial purE downstream metabolite 5-aminoimidazole-4-carboxamide ribotide (AICAR) can inhibit worm growth. Additionally, we found the FoxO transcription factor DAF-16 is indispensable in worm development delay induced by purE mutation, and observed increased nuclear accumulation of DAF-16 when fed E. coli purE- mutants, suggesting the role of DAF-16 in response to purE mutation. RNA-seq analysis and phenotypic assays revealed that worms fed the E. coli purE mutant exhibited elevated lifespan, thermotolerance, and pathogen resistance. These findings collectively suggest that certain intermediates in the bacterial purine biosynthesis can serve as a cue to modulate development and activate the defense response in the nematode C. elegans through DAF-16.
Collapse
Affiliation(s)
- Min Feng
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Baizhen Gao
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - L Rene Garcia
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Qing Sun
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| |
Collapse
|
54
|
Fanfarillo F, Caronti B, Lucarelli M, Francati S, Tarani L, Ceccanti M, Piccioni MG, Verdone L, Caserta M, Venditti S, Ferraguti G, Fiore M. Alcohol Consumption and Breast and Ovarian Cancer Development: Molecular Pathways and Mechanisms. Curr Issues Mol Biol 2024; 46:14438-14452. [PMID: 39727994 DOI: 10.3390/cimb46120866] [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: 11/20/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
Alcohol consumption has been consistently linked to an increased risk of several cancers, including breast and ovarian cancer. Despite substantial evidence supporting this association, the precise mechanisms underlying alcohol's contribution to cancer pathogenesis remain incompletely understood. This narrative review focuses on the key current literature on the biological pathways through which alcohol may influence the development of breast and ovarian cancer. Key mechanisms discussed include the modulation of estrogen levels, the generation of reactive oxygen species, the production of acetaldehyde, the promotion of chronic inflammation, and the induction of epigenetic changes. Alcohol's impact on estrogenic signaling, particularly in the regulation of estrogen and progesterone, is explored in the context of hormone-dependent cancers. Additionally, the role of alcohol-induced DNA damage, mutagenesis, and immune system modulation in tumor initiation and progression is examined. Overall, this review emphasizes the importance of alcohol as a modifiable risk factor for breast and ovarian cancer and highlights the need for further research to clarify its role in cancer biology.
Collapse
Affiliation(s)
- Francesca Fanfarillo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Brunella Caronti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Lucarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Silvia Francati
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Luigi Tarani
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Mauro Ceccanti
- SITAC, Società Italiana per il Trattamento dell'Alcolismo e le sue Complicanze, 00185 Rome, Italy
| | - Maria Grazia Piccioni
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Loredana Verdone
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy
| | - Micaela Caserta
- Institute of Molecular Biology and Pathology (IBPM-CNR), 00161 Rome, Italy
| | - Sabrina Venditti
- Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00161 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
55
|
Matsumoto K, Akieda Y, Haraoka Y, Hirono N, Sasaki H, Ishitani T. Foxo3-mediated physiological cell competition ensures robust tissue patterning throughout vertebrate development. Nat Commun 2024; 15:10662. [PMID: 39690179 PMCID: PMC11652645 DOI: 10.1038/s41467-024-55108-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Unfit cells with defective signalling or gene expression are eliminated through competition with neighbouring cells. However, physiological roles and mechanisms of cell competition in vertebrates remain unclear. In addition, universal mechanisms regulating diverse cell competition are unknown. Using zebrafish imaging, we reveal that cell competition ensures robust patterning of the spinal cord and muscle through elimination of cells with unfit sonic hedgehog activity, driven by cadherin-mediated communication between unfit and neighbouring fit cells and subsequent activation of the Smad-Foxo3-reactive oxygen species axis. We identify Foxo3 as a common marker of loser cells in various types of cell competition in zebrafish and mice. Foxo3-mediated physiological cell competition is required for eliminating various naturally generated unfit cells and for the consequent precise patterning during zebrafish embryogenesis and organogenesis. Given the implication of Foxo3 downregulation in age-related diseases, cell competition may be a defence system to prevent abnormalities throughout development and adult homeostasis.
Collapse
Affiliation(s)
- Kanako Matsumoto
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Suita, Osaka, Japan
| | - Yuki Akieda
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yukinari Haraoka
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Naoki Hirono
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Sasaki
- Laboratory for Embryogenesis, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | - Tohru Ishitani
- Department of Homeostatic Regulation, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.
- Department of Biological Sciences, Graduate School of Science, Osaka University, Suita, Osaka, Japan.
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka, Japan.
| |
Collapse
|
56
|
Sementino E, Hassan D, Bellacosa A, Testa JR. AKT and the Hallmarks of Cancer. Cancer Res 2024; 84:4126-4139. [PMID: 39437156 DOI: 10.1158/0008-5472.can-24-1846] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/17/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Nearly a quarter century ago, Hanahan and Weinberg conceived six unifying principles explaining how normal cells transform into malignant tumors. Their provisional set of biological capabilities acquired during tumor development-cancer hallmarks-would evolve to 14 tenets as knowledge of cancer genomes, molecular mechanisms, and the tumor microenvironment expanded, most recently adding four emerging enabling characteristics: phenotypic plasticity, epigenetic reprogramming, polymorphic microbiomes, and senescent cells. AKT kinases are critical signaling molecules that regulate cellular physiology upon receptor tyrosine kinases and PI3K activation. The complex branching of the AKT signaling network involves several critical downstream nodes that significantly magnify its functional impact, such that nearly every organ system and cell in the body may be affected by AKT activity. Conversely, tumor-intrinsic dysregulation of AKT can have numerous adverse cellular and pathologic ramifications, particularly in oncogenesis, as multiple tumor suppressors and oncogenic proteins regulate AKT signaling. Herein, we review the mounting evidence implicating the AKT pathway in the aggregate of currently recognized hallmarks of cancer underlying the complexities of human malignant diseases. The challenges, recent successes, and likely areas for exciting future advances in targeting this complex pathway are also discussed.
Collapse
Affiliation(s)
- Eleonora Sementino
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Dalal Hassan
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
- Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Alfonso Bellacosa
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Joseph R Testa
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
57
|
Masedunskas A, de Ciutiis I, Hein LK, Ge A, Kong YX, Qi M, Mainali D, Rogerson-Wood L, Kroeger CM, Aguirre Candia YA, Cagigas ML, Wang T, Hutchinson D, Sabag A, Passam FH, Piccio L, Sargeant TJ, Fontana L. Investigating the Impact of Glycogen-Depleting Exercise Combined with Prolonged Fasting on Autophagy and Cellular Health in Humans: A Randomised Controlled Crossover Trial. Nutrients 2024; 16:4297. [PMID: 39770918 PMCID: PMC11677747 DOI: 10.3390/nu16244297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
IMPORTANCE Although prolonged fasting has become increasingly popular, the favourable biological adaptations and possible adverse effects in humans have yet to be fully elucidated. OBJECTIVE To investigate the effects of a three-day water-only fasting, with or without exercise-induced glycogen depletion, on autophagy activation and the molecular pathways involved in cellular damage accumulation and repair in healthy humans. DESIGN A randomised, single-centre, two-period, two-sequence crossover trial. The primary outcome is autophagic activity, assessed as flux in peripheral blood mononuclear cells (PBMCs) measured in the context of whole blood. Secondary outcomes include changes in body composition, heart rate variability, endothelial function, and genomic, epigenomic, metabolomic, proteomic, and metagenomic adaptations to fasting in plasma, platelets, urine, stools, and PBMCs. Detailed profiling of circulating immune cell populations and their functional states will be assessed by flow cytometry. SETTING All clinical investigations will be undertaken at the Charles Perkins Centre Royal Prince Alfred Hospital clinic, University of Sydney, Australia. PARTICIPANTS Twenty-four individuals aged 18 to 70 years, with a BMI of 20-40 kg/m2, free of major health conditions other than obesity. DISCUSSION While autophagic flux induction through fasting has garnered interest, there is a notable lack of human studies on this topic. This trial aims to provide the most detailed and integrated analysis of how three days of prolonged water-only fasting, combined with glycogen-depleting exercise, affects autophagy activation and other crucial metabolic and molecular pathways linked to cellular, metabolic, and immune health. Insights from this study may pave the way for safe and effective strategies to induce autophagy, offering potential preventive interventions for a range of chronic conditions.
Collapse
Affiliation(s)
- Andrius Masedunskas
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Isabella de Ciutiis
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Leanne K. Hein
- Lysosomal Health in Ageing, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
| | - Anjie Ge
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Yvonne X. Kong
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Miao Qi
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Drishya Mainali
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Lara Rogerson-Wood
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Cynthia M. Kroeger
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Yvonne A. Aguirre Candia
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Maria L. Cagigas
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Tian Wang
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - David Hutchinson
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia
| | - Angelo Sabag
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Freda H. Passam
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Laura Piccio
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
| | - Timothy J. Sargeant
- Lysosomal Health in Ageing, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Luigi Fontana
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; (A.M.); (A.G.); (A.S.); (F.H.P.)
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia
| |
Collapse
|
58
|
Butucescu M, Imre M, Rus-Hrincu F, Voicu-Balasea B, Popa A, Moisa M, Ripszky A, Neculau C, Pituru SM, Pârvu S. Cell-Type-Specific ROS-AKT/mTOR-Autophagy Interplay-Should It Be Addressed in Periimplantitis? Diagnostics (Basel) 2024; 14:2784. [PMID: 39767145 PMCID: PMC11727345 DOI: 10.3390/diagnostics14242784] [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: 11/04/2024] [Accepted: 12/09/2024] [Indexed: 01/03/2025] Open
Abstract
Periimplantitis represents an inflammatory disease of the soft and hard tissues surrounding the osseointegrated dental implant, triggering progressive damage to the alveolar bone. Cumulative data have revealed that periimplantitis plays a crucial part in implant failure. Due to the strategic roles of autophagy and its upstream coordinator, the AKT/mTOR pathway, in inflammatory responses, the crosstalk between them in the context of periimplantitis should become a key research target, as it opens up an area of interesting data with clinical significance. Therefore, in this article, we aimed to briefly review the existing data concerning the complex roles played by ROS in the interplay between the AKT/mTOR signaling pathway and autophagy in periimplantitis, in each of the main cell types involved in periimplantitis pathogenesis and evolution. Knowing how to modulate specifically the autophagic machinery in each of the cellular types involved in the healing and osseointegration steps post implant surgery can help the clinician to make the most appropriate post-surgery decisions. These decisions might be crucial in order to prevent the occurrence of periimplantitis and ensure the proper conditions for effective osseointegration, depending on patients' clinical particularities.
Collapse
Affiliation(s)
- Mihai Butucescu
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Marina Imre
- Department of Prosthodontics, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Calea Plevnei, 010221 Bucharest, Romania;
| | - Florentina Rus-Hrincu
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Bianca Voicu-Balasea
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Alexandra Popa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Mihai Moisa
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
| | - Alexandra Ripszky
- Department of Biochemistry, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania; (F.R.-H.); (A.P.); (M.M.); (A.R.)
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Cristina Neculau
- The Interdisciplinary Center for Dental Research and Development, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Silviu Mirel Pituru
- Department of Organization, Professional Legislation and Management of the Dental Office, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 17-23 Plevnei Street, 020021 Bucharest, Romania;
| | - Simona Pârvu
- National Institute of Public Health, General Medicine Faculty, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
59
|
Willis BS, Mongeon K, Dry H, Neveras IL, Bryan N, Pandya M, Roderick-Richardson J, Xu W, Yang L, Rosen A, Reimer C, Tuskova L, Klener P, Mettetal JT, Lenz G, Barry ST. Potent combination benefit of the AKT inhibitor capivasertib and the BCL-2 inhibitor venetoclax in diffuse large B cell lymphoma. Leukemia 2024; 38:2663-2674. [PMID: 39284898 PMCID: PMC11588655 DOI: 10.1038/s41375-024-02401-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/28/2024] [Indexed: 11/27/2024]
Abstract
The therapeutic potential of targeting PI3K/AKT/PTEN signalling in B-cell malignancies remains attractive. Whilst PI3K-α/δ inhibitors demonstrate clinical benefit in certain B-cell lymphomas, PI3K signalling inhibitors have been inadequate in relapsed/refractory diffuse large B-cell lymphoma (DLBCL) in part, due to treatment related toxicities. Clinically, AKT inhibitors exhibit a differentiated tolerability profile offering an alternative approach for treating patients with B-cell malignancies. To explore how AKT inhibition complements other potential therapeutics in the treatment of DLBCL patients, an in vitro combination screen was conducted across a panel of DLCBL cell lines. The AKT inhibitor, capivasertib, in combination with the BCL-2 inhibitor, venetoclax, produced notable therapeutic benefit in preclinical models of DLBCL. Capivasertib and venetoclax rapidly induced caspase and PARP cleavage in GCB-DLBCL PTEN wildtype cell lines and those harbouring PTEN mutations or reduced PTEN protein, driving prolonged tumour growth inhibition in DLBCL cell line and patient derived xenograft lymphoma models. The addition of the rituximab further deepened the durability of capivasertib and venetoclax responses in a RCHOP refractory DLBCL in vivo models. These findings provide preclinical evidence for the rational treatment combination of AKT and BCL-2 inhibitors using capivasertib and venetoclax respectively alongside anti-CD20 antibody supplementation for treatment of patients with DLBCL.
Collapse
Affiliation(s)
| | - Kevin Mongeon
- Bioscience, Early Oncology, AstraZeneca, Boston, USA
| | - Hannah Dry
- Bioscience, Early Oncology, AstraZeneca, Boston, USA
| | | | - Nadezda Bryan
- Bioscience, Early Oncology, AstraZeneca, Boston, USA
| | | | | | - Wendan Xu
- Department of Medicine A, Haematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Li Yang
- Department of Medicine A, Haematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Alan Rosen
- Bioscience, Early Oncology, AstraZeneca, Boston, USA
| | | | - Liliana Tuskova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University Prague, Prague, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University Prague, Prague, Czech Republic
| | | | - Georg Lenz
- Department of Medicine A, Haematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK.
| |
Collapse
|
60
|
Enomoto S, Nakatsuka S, Kuwayama T, Kawatsu K, Yokogawa M, Osawa M. NMR 1H, 13C, 15N backbone resonance assignments of 14-3-3ζ binding region of human FOXO3a (residues 1-284). BIOMOLECULAR NMR ASSIGNMENTS 2024; 18:275-283. [PMID: 39259470 PMCID: PMC11511766 DOI: 10.1007/s12104-024-10200-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
In tumors, mutation in Ras proteins stimulates a signaling cascade through phosphorylation. Downstream of the cascade, many transcription and translation factors are up- or down-regulated by phosphorylation, leading to cancer progression. This phosphorylation cascade is sustained by 14-3-3ζ protein. 14-3-3ζ binds to its client proteins that are Ser/Thr-phosphorylated and prevents their dephosphorylation. One of those transcription factors is FOXO3a, whose transcriptional activity is suppressed in the phosphorylation cascade. FOXO3a binds to specific DNA sequences and activates the transcription of apoptosis-related proteins. In cancer cells, however, FOXO3a is phosphorylated, bound to 14-3-3ζ, and dissociated from the DNA, resulting in FOXO3a inactivation. To elucidate the mechanism of FOXO3a inactivation by the 14-3-3ζ binding, we aim to perform NMR analysis of the interaction between 14-3-3ζ and di-phosphorylated FOXO3a residues 1-284 (dpFOXO3a). Here, we report the backbone resonance assignments of dpFOXO3a, which are transferred from those of the N-terminal domain (NTD) and the DNA-binding domain (DBD) of dpFOXO3a.
Collapse
Affiliation(s)
- Shota Enomoto
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Shoichi Nakatsuka
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Tomoya Kuwayama
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Kosaku Kawatsu
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Mariko Yokogawa
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan
| | - Masanori Osawa
- Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo, Japan.
| |
Collapse
|
61
|
Diggins NL, Pham AH, Mitchell J, Parkins CJ, Slind L, Turner R, Lee BJ, Yurochko AD, Caposio P, Nelson JA, Hancock MH. Viral microRNA regulation of Akt is necessary for reactivation of Human Cytomegalovirus from latency in CD34+ hematopoietic progenitor cells and humanized mice. PLoS Pathog 2024; 20:e1012285. [PMID: 39661658 PMCID: PMC11666035 DOI: 10.1371/journal.ppat.1012285] [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: 05/23/2024] [Revised: 12/23/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Human cytomegalovirus (HCMV) actively manipulates cellular signaling pathways to benefit viral replication. Phosphatidyl-inositol 3-kinase (PI3K)/Akt signaling is an important negative regulator of HCMV replication, and during lytic infection the virus utilizes pUL38 to limit Akt phosphorylation and activity. During latency, PI3K/Akt signaling also limits virus replication, but how this is overcome at the time of reactivation is unknown. Virally encoded microRNAs (miRNAs) are a key component of the virus arsenal used to alter signaling during latency and reactivation. In the present study we show that three HCMV miRNAs (miR-UL36, miR-UL112 and miR-UL148D) downregulate Akt expression and attenuate downstream signaling, resulting in the activation of FOXO3a and enhanced internal promoter-driven IE transcription. A virus lacking expression of all three miRNAs is unable to reactivate from latency both in CD34+ hematopoietic progenitor cells and in a humanized mouse model of HCMV infection, however downregulating Akt restores the ability of the mutant virus to replicate. These findings highlight the negative role Akt signaling plays in HCMV replication in lytic and latent infection and how the virus has evolved miRNA-mediated countermeasures to promote successful reactivation.
Collapse
Affiliation(s)
- Nicole L. Diggins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Andrew H. Pham
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jennifer Mitchell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Luke Slind
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Rebekah Turner
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Byeong-Jae Lee
- Department of Microbiology & Immunology, Center for Applied Immunology and Pathological Processes, Center for Emerging Viral Threats, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Andrew D. Yurochko
- Department of Microbiology & Immunology, Center for Applied Immunology and Pathological Processes, Center for Emerging Viral Threats, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jay A. Nelson
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Meaghan H. Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
62
|
Korb J. Termites and other social insects as emerging model organisms of ageing research: how to achieve a long lifespan and a high fecundity. J Exp Biol 2024; 227:jeb246497. [PMID: 39535049 DOI: 10.1242/jeb.246497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Social insects (termites, ants and some bees and wasps) are emerging model organisms of ageing research. In this Commentary, I outline which advantages they offer compared with other organisms. These include the co-occurrence of extraordinarily long-lived, highly fecund queens together with short-lived workers within colonies that share the same genetic background. I then summarize which new insights have been gained so far from social insect studies. Research on social insects has led to the development of a universal mechanistic framework underlying the regulation of ageing and other life-history trade-offs in insects: the TI-J-LiFe network (short for TOR/IIS-juvenile hormone-lifespan/fecundity). Because of its conservative nature, this network can be extended to also incorporate vertebrates. Current data for social insect models suggest that molecular re-wirings along the I-J-Fe (IIS-juvenile hormone-fecundity) axis of the network can explain the concurrent long lifespans and high fecundity of queens. During social evolution, pathways that foster a high fecundity have apparently been uncoupled from mechanisms that shorten lifespan in solitary insects. Thus, fecundity-related vitellogenesis is uncoupled from life-shortening high juvenile hormone (JH)-titres in the honeybee and from insulin/insulin-like growth factor signalling (IIS) activity in ants. In termites, similarly, vitellogenesis seems tissue-specifically unlinked from JH signalling and IIS activity might have lost life-shortening consequences. However, as in solitary animals, the downstream processes (Li of the TI-J-LiFe network) that cause actual ageing (e.g. oxidative stress, transposable element activity, telomere attrition) seem to differ between species and environments. These results show how apparently hard-wired mechanisms underlying life-history trade-offs can be overcome during evolution.
Collapse
Affiliation(s)
- Judith Korb
- Evolutionary Biology & Ecology, University of Freiburg, D-79104 Freiburg, Germany
- Research Institute for the Environment and Livelihoods, Charles Darwin University, Darwin, NT 0810, Australia
| |
Collapse
|
63
|
Shan XQ, Zhou N, Pei CX, Lu X, Chen CP, Chen HQ. Tetrandrine induces muscle atrophy involving ROS-mediated inhibition of Akt and FoxO3. Mol Med 2024; 30:218. [PMID: 39548359 PMCID: PMC11566300 DOI: 10.1186/s10020-024-00981-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Tetrandrine (Tet), a well-known drug of calcium channel blocker, has been broadly applied for anti-inflammatory and anti-fibrogenetic therapy. However, due to the functional diversity of ubiquitous calcium channels, potential side-effects may be expected. Our previous report revealed an inhibitory effect of Tet on myogenesis of skeletal muscle. Here, we found that Tet induced protein degradation resulting in the myofibril atrophy. Upon administration with a relative high dose (40 mg/kg) of Tet for 28 days, the mice displayed significantly reduced muscle mass, strength force, and myosin heavy chain (MyHC) protein levels. The MyHC reduction was further detected in C2C12 myotubes after treating with Tet. Interestingly, the expression of Atrogin-1 and Murf-1, the skeletal muscle specific E3 ligases of protein ubiquitin-proteasome system (UPS), was accordingly up-regulated, and the reduced MyHC was significantly mitigated by MG132, a 26S proteasome inhibitor, indicating a key role of UPS in the protein degradation of muscle cells. Further study showed that Tet induced autophagy also participated in the protein degradation. Mechanistically, Tet treatment caused ROS production in myotubes that in turn targeted on FoxO3/AKT signaling, resulting in the activation of UPS and autophagy processes that were involved in the protein degradation. Our study reveals a potential side-effect of Tet on skeletal muscle atrophy, particularly when the drug dose is relatively high.
Collapse
Affiliation(s)
- Xin-Qi Shan
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Na Zhou
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Chuang-Xin Pei
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Xue Lu
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Cai-Ping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| | - Hua-Qun Chen
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
64
|
Kamayirese S, Hansen LA, Lovas S. Negative Charges, Not Necessary Phosphorylation, are Required for Ligand Recognition by 14-3-3 Proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613320. [PMID: 39345434 PMCID: PMC11429721 DOI: 10.1101/2024.09.16.613320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Protein-protein interactions involving 14-3-3 proteins regulate various cellular activities in normal and pathological conditions. These interactions have mostly been reported to be phosphorylation-dependent, but the 14-3-3 proteins also interact with unphosphorylated proteins. In this work, we investigated whether phosphorylation is required, or, alternatively, whether negative charges are sufficient for 14-3-3ε binding. We substituted the pThr residue of pT(502-510) peptide by residues with varying number of negative charges, and investigated binding of the peptides to 14-3-3ε using MD simulations and biophysical methods. We demonstrated that at least one negative charge is required for the peptides to bind 14-3-3ε while phosphorylation is not necessary, and that two negative charges are preferable for high affinity binding.
Collapse
Affiliation(s)
- Seraphine Kamayirese
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| | - Laura A. Hansen
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| | - Sándor Lovas
- Department of Biomedical Sciences, Creighton University, Omaha, Nebraska 68178, United States
| |
Collapse
|
65
|
Hlavac K, Pavelkova P, Ondrisova L, Mraz M. FoxO1 signaling in B cell malignancies and its therapeutic targeting. FEBS Lett 2024. [PMID: 39533662 DOI: 10.1002/1873-3468.15057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
FoxO transcription factors (FoxO1, FoxO3a, FoxO4, FoxO6) are a highly evolutionary conserved subfamily of the 'forkhead' box proteins. They have traditionally been considered tumor suppressors, but FoxO1 also exhibits oncogenic properties. The complex nature of FoxO1 is illustrated by its various roles in B cell development and differentiation, immunoglobulin gene rearrangement and cell-surface B cell receptor (BCR) structure, DNA damage control, cell cycle regulation, and germinal center reaction. FoxO1 is tightly regulated at a transcriptional (STAT3, HEB, EBF, FoxOs) and post-transcriptional level (Akt, AMPK, CDK2, GSK3, IKKs, JNK, MAPK/Erk, SGK1, miRNA). In B cell malignancies, recurrent FoxO1 activating mutations (S22/T24) and aberrant nuclear export and activity have been described, underscoring the potential of its therapeutic inhibition. Here, we review FoxO1's roles across B cell and myeloid malignancies, namely acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), follicular lymphoma (FL), diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL), Burkitt lymphoma (BL), Hodgkin lymphoma (HL), and multiple myeloma (MM). We also discuss preclinical evidence for FoxO1 targeting by currently available inhibitors (AS1708727, AS1842856, cpd10).
Collapse
Affiliation(s)
- Krystof Hlavac
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Petra Pavelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Laura Ondrisova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| | - Marek Mraz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
66
|
Cao J, Chen S, Wang J, Fan X, Liu S, Li X, Yang L. Transcription factor PRRX1-activated ANXA6 facilitates EGFR-PKCα complex formation and enhances cisplatin sensitivity in bladder cancer. Life Sci 2024; 359:123228. [PMID: 39528080 DOI: 10.1016/j.lfs.2024.123228] [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: 07/10/2024] [Revised: 10/20/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Tumor resistance to cisplatin represents a major clinical challenge, particularly in bladder cancer (BC). ANXA6 is a member of annexin family, and its role in cisplatin resistance remains unclear. This study explores ANXA6's role in promoting cisplatin sensitivity. METHODS Bioinformatics analyses and clinical specimen verifications assessed the correlation between ANXA6 and cisplatin treatment. A series of assays, including CCK-8, clone formation assay, flow cytometry assays for reactive oxygen species (ROS) and apoptosis, and comet assays, were used to confirm ANXA6's role in enhancing cisplatin sensitivity and re-sensitizing resistant BC cells. Mass spectrometry, immunofluorescence, and co-immunoprecipitation experiments elucidated ANXA6's role in enhancing PKCα/EGFR complex formation and inhibiting the EGFR pathway. ChIP-PCR and dual-luciferase assays determined PRRX1's regulatory role on ANXA6 transcription. Finally, the impact of ANXA6 in vivo was evaluated using xenograft models. RESULTS Bioinformatics analyses showed a significant correlation between ANXA6 expression and cisplatin sensitivity. In vitro and in vivo experiments confirmed that ANXA6 was a new target for cisplatin treatment. ANXA6 overexpression not only enhanced cell viability inhibition, DNA damage and apoptosis caused by cisplatin, but also re-sensitized cisplatin-resistant cells. Mechanistically, ANXA6 promotes PKCα/EGFR complex formation, inhibiting EGFR phosphorylation and downstream AKT and ERK1/2. Moreover, PRRX1 was identified as a transcription factor promoting ANXA6 expression, thereby augmenting the cytotoxic effects of cisplatin. CONCLUSION Our study reveals the mechanism by which ANXA6 enhances cisplatin sensitivity and re-sensitizes resistant cells. The roles of PRRX1 and ANXA6 in cisplatin resistance offer new therapeutic targets to overcome cisplatin resistance in clinical practice.
Collapse
Affiliation(s)
- Jinlong Cao
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China
| | - Siyu Chen
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China
| | - Jirong Wang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China
| | - Xinpeng Fan
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China
| | - Shanhui Liu
- Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China
| | - Xiaoran Li
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China.
| | - Li Yang
- Department of Urology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China; Gansu Province Clinical Research Center for Urology, Lanzhou 730000, China.
| |
Collapse
|
67
|
Kokot T, Zimmermann JP, Schwäble AN, Reimann L, Herr AL, Höfflin N, Köhn M, Warscheid B. Protein phosphatase-1 regulates the binding of filamin C to FILIP1 in cultured skeletal muscle cells under mechanical stress. Sci Rep 2024; 14:27348. [PMID: 39521905 PMCID: PMC11550807 DOI: 10.1038/s41598-024-78953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The actin-binding protein filamin c (FLNc) is a key mediator in the response of skeletal muscle cells to mechanical stress. In addition to its function as a structural scaffold, FLNc acts as a signaling adaptor which is phosphorylated at S2234 in its mechanosensitive domain 20 (d20) through AKT. Here, we discovered a strong dephosphorylation of FLNc-pS2234 in cultured skeletal myotubes under acute mechanical stress, despite high AKT activity. We found that all three protein phosphatase 1 (PP1) isoforms are part of the FLNc d18-21 interactome. Enzymatic assays demonstrate that PP1 efficiently dephosphorylates FLNc-pS2234 and in vitro and in cells upon PP1 activation using specific modulators. FLNc-pS2234 dephosphorylation promotes the interaction with FILIP1, a mediator for filamin degradation. Altogether, we present a model in which dephosphorylation of FLNc d20 by the dominant action of PP1c prevails over AKT activity to promote the binding of the filamin degradation-inducing factor FILIP1 during acute mechanical stress.
Collapse
Affiliation(s)
- Thomas Kokot
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Johannes P Zimmermann
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Anja N Schwäble
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Lena Reimann
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Anna L Herr
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Sartorius CellGenix GmbH, Freiburg, Germany
| | - Nico Höfflin
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Maja Köhn
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Bettina Warscheid
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany.
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
68
|
Xuan R, Hu T, Cai L, Zhao B, Han E, Xia Z. CARD16 restores tumorigenesis and restraints apoptosis in glioma cells Via FOXO1/TRAIL axis. Cell Death Dis 2024; 15:804. [PMID: 39516471 PMCID: PMC11549220 DOI: 10.1038/s41419-024-07196-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
A hallmark of glioma cells, particularly glioblastoma multiforme (GBM) cells, is their resistance to apoptosis. Accumulating evidences has demonstrated that CARD16, a caspase recruitment domain (CARD) only protein, enhances both anti-apoptotic and tumorigenic properties. Nevertheless, there is a limited understanding of the expression and functional role of CARD16 in glioma. This study seeks to investigate, through in silico analysis and clinical specimens, the role of CARD16 as a potential tumor promoter in glioma. Functional assays and molecular studies revealed that CARD16 promotes tumorigenesis and suppresses apoptosis in glioma cells. Moreover, knockdown of CARD16 enhances the expression of the FOXO1/TRAIL axis in GBM cells. Additionally, FOXO1 downregulation in CARD16 knockdown GBM cells restores proliferation and reduces apoptosis. Further investigation demonstrated that elevated P21 expression inhibits CDK2-mediated FOXO1 phosphorylation and ubiquitination in CARD16-knockdown GBM cells. Collectively, these findings suggest that CARD16 is a tumor-promoting molecular in glioma via downregulating FOXO1/TRAIL axis, and suppressing TRAIL-induced apoptosis. The CARD16 gene presents significant potential for prognostic prediction and advances in innovative apoptotic therapeutics.
Collapse
Affiliation(s)
- Ruoheng Xuan
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tianyu Hu
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lingshan Cai
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Beichuan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Erqiao Han
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhibo Xia
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
69
|
Shafiek MS, Mekky RY, Nassar NN, El-Yamany MF, Rabie MA. Vortioxetine ameliorates experimental autoimmune encephalomyelitis model of multiple sclerosis in mice via activation of PI3K/Akt/CREB/BDNF cascade and modulation of serotonergic pathway signaling. Eur J Pharmacol 2024; 982:176929. [PMID: 39181226 DOI: 10.1016/j.ejphar.2024.176929] [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: 05/22/2024] [Revised: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition characterized by immune cell infiltration and cytokine overproduction that led to myelin sheath inflammatory assaults, thus causing axonal destruction. The former consequently provokes motor impairment and psychological disorders. Markedly, depression is one of the most prevalent lifelong comorbidities that negatively impacts the quality of life in MS patients. Vortioxetine (VTX), a multi-modal molecule prescribed to manage depression and anxiety disorder, additionally, it displays a promising neuroprotective properties against neurodegenerative diseases such as Alzheimer's and Parkinson's. To this end, the present study investigated the potential therapeutic efficacy of VTX against experimental autoimmune encephalomyelitis (EAE) model of MS in mice. Notably, treatment with VTX significantly ameliorated EAE-induced motor disability, as evident by enhanced performance in open field, rotarod and grip strength tests, alongside a reduction in immobility time during the forced swimming test, indicating a mitigation of the depressive-like behavior; outcomes that were corroborated with histological examinations and biochemical analyses. Mechanistically, VTX enhanced serotonin levels by inhibiting both serotonin transporter (SERT) and indoleamine 2,3-dioxygenase (IDO) enzyme, thereby promoting the activation of serotonin 1A (5-HT1A) receptor. The latter triggered the stimulation of phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) cascade that entailed activation/phosphorylation of cAMP response element-binding protein (CREB). This activation increased brain derived neurotrophic factor (BDNF) and myelin basic protein (MBP) contents that mitigated demyelination in the corpus callosum. Furthermore, VTX suppressed phospho serine 536 nuclear factor kappa B (pS536 NF-κB p65) activity and reduced tumor necrosis factor-alpha (TNF-α) production. The results underscore VTX's beneficial effects on disease severity in EAE model of MS in mice by amending both inflammatory and neurodegenerative components of MS progression.
Collapse
Affiliation(s)
- Marwa S Shafiek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | - Radwa Y Mekky
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt
| | - Noha N Nassar
- Department of Pharmacology and Toxicology, October University for Modern Science and Arts (MSA), Giza, 12622, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| |
Collapse
|
70
|
Zhang H, Gao H, Lin X, Yang B, Wang J, Yuan X, Zhang Z, He T, Liu Z. Akt-FoxO signaling drives co-adaptation to insecticide and host plant stresses in an herbivorous insect. J Adv Res 2024:S2090-1232(24)00498-3. [PMID: 39510378 DOI: 10.1016/j.jare.2024.11.006] [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: 07/21/2024] [Revised: 10/07/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024] Open
Abstract
INTRODUCTION Ongoing interactions between host and herbivorous insect trigger a co-evolutionary arms race. Genetic diversity within insects facilitates their adaptation to phytochemicals and their derivatives, including plant-derived insecticides. Cytochrome P450s play important roles in metabolizing phytochemicals and insecticides, due to their diversity and almost perfect evolution. OBJECTIVES This study aims to uncover a common molecular mechanism in herbivorous insects by investigating the role of kinase-transcription factor regulation of P450s in conferring tolerance to both insecticides and phytochemicals. METHODS RNA interference, transcriptome sequencing, insecticide, and phytochemical bioassays were conducted to validate the functions of Akt, FoxO, and candidate P450s. Dual-luciferase activity assays were employed to identify the regulation of P450s by the Akt-FoxO signaling pathway. Recombinant P450 enzymes were utilized to investigate the metabolism of insecticides and phytochemicals. RESULTS We identified an Akt-FoxO signaling cascade, a representative of kinase-transcription factor pathways. This cascade mediates the expression of eight P450 enzymes involved in the metabolism of insecticides and phytochemicals in Nilaparvata lugens. These P450s are from different families and with different substrate selectivity, enabling them to respectively metabolize insecticides and phytochemicals with structure diversity. Nevertheless, the eight P450 genes were up-regulated by FoxO, which was inhibited in a higher cascade by Akt through phosphorylation. The discovery of the Akt-FoxO signaling pathway regulating a series of P450 genes elucidates the finely tuned regulatory mechanism in insects for adapting to phytochemicals and insecticides. CONCLUSION These finding sheds light on the physiological balance maintained by these regulatory processes. The work provides the experimental evidence of co-adaptation to the stresses imposed by host plant and insecticide within the model of the kinase-TF involving various P450s. This model provides a comprehensive view of how pests adapt to multiple environmental stresses.
Collapse
Affiliation(s)
- Huihui Zhang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Haoli Gao
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Xumin Lin
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Baojun Yang
- Rice Technology Research and Development Center, China National Rice Research Institute, Stadium 359, Hangzhou 310006, China
| | - Jingting Wang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Xiaowei Yuan
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Zhen Zhang
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Tianshun He
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Zewen Liu
- Key Laboratory of Integrated Management of Crop Diseases and Pests (Ministry of Education), College of Plant Protection, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China.
| |
Collapse
|
71
|
Golec DP, Gazzinelli-Guimaraes P, Chauss D, Nagashima H, Yu K, Hill T, Nivelo L, Cannons JL, Perry J, Joshi I, Pereira N, Oliveira FMS, Cruz AC, Druey KM, Lack JB, Nutman TB, Villarino AV, O'Shea JJ, Afzali B, Schwartzberg PL. A PI3Kδ-Foxo1-FasL signaling amplification loop rewires CD4 + T helper cell signaling, differentiation and epigenetic remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620691. [PMID: 39803425 PMCID: PMC11722529 DOI: 10.1101/2024.10.28.620691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
While inputs regulating CD4+ T helper cell (Th) differentiation are well-defined, the integration of downstream signaling with transcriptional and epigenetic programs that define Th-lineage identity remain unresolved. PI3K signaling is a critical regulator of T cell function; activating mutations affecting PI3Kδ result in an immunodeficiency with multiple T cell defects. Using mice expressing activated-PI3Kδ, we found aberrant expression of proinflammatory Th1-signature genes under Th2-inducing conditions, both in vivo and in vitro. This dysregulation was driven by a robust PI3Kδ-IL-2-Foxo1 signaling loop, fueling Foxo1-inactivation, loss of Th2-lineage restriction, altered chromatin accessibility and global impairment of CTCF-DNA interactions. Surprisingly, ablation of Fasl, a Foxo1-repressed gene, restored normal Th2 differentiation, TCR signaling and CTCF expression. BioID revealed Fas interactions with TCR-signaling components, which were supported by Fas-mediated potentiation of TCR signaling. Our results highlight Fas-FasL signaling as a critical intermediate in phenotypes driven by activated-PI3Kδ, thereby linking two key pathways of immune dysregulation.
Collapse
Affiliation(s)
- Dominic P Golec
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Pedro Gazzinelli-Guimaraes
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Science, George Washington University, Washington, DC, USA
| | - Daniel Chauss
- Immunoregulation Section, NIDDK, NIH, Bethesda, MD, USA
| | | | - Kang Yu
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Tom Hill
- NIAID Collaborative Bioinformatics Resource (NCBR), NIAID, NIH, Bethesda, MD, USA
| | - Luis Nivelo
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Jillian Perry
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Ilin Joshi
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Nicolas Pereira
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Fabrício Marcus Silva Oliveira
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Science, George Washington University, Washington, DC, USA
| | - Anthony C Cruz
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Kirk M Druey
- Laboratory of Allergic Diseases, NIAID, NIH, Bethesda, Maryland, USA
| | - Justin B Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), NIAID, NIH, Bethesda, MD, USA
| | - Thomas B Nutman
- Laboratory of Parasitic Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Alejandro V Villarino
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - John J O'Shea
- Lymphocyte Cell Biology Section, NIAMS, NIH, Bethesda, MD, USA
| | - Behdad Afzali
- Immunoregulation Section, NIDDK, NIH, Bethesda, MD, USA
| | | |
Collapse
|
72
|
Rautela U, Sarkar GC, Chaudhary A, Chatterjee D, Rosh M, Arimbasseri AG, Mukhopadhyay A. A non-canonical role of somatic Cyclin D/CYD-1 in oogenesis and in maintenance of reproductive fidelity, dependent on the FOXO/DAF-16 activation state. PLoS Genet 2024; 20:e1011453. [PMID: 39546504 PMCID: PMC11602045 DOI: 10.1371/journal.pgen.1011453] [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: 06/10/2024] [Revised: 11/27/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024] Open
Abstract
For the optimal survival of a species, an organism coordinates its reproductive decisions with the nutrient availability of its niche. Thus, nutrient-sensing pathways like insulin-IGF-1 signaling (IIS) play an important role in modulating cell division, oogenesis, and reproductive aging. Lowering of the IIS leads to the activation of the downstream FOXO transcription factor (TF) DAF-16 in Caenorhabditis elegans which promotes oocyte quality and delays reproductive aging. However, less is known about how the IIS axis responds to changes in cell cycle proteins, particularly in the somatic tissues. Here, we show a new aspect of the regulation of the germline by this nutrient-sensing axis. First, we show that the canonical G1-S cyclin, Cyclin D/CYD-1, regulates reproductive fidelity from the uterine tissue of wild-type worms. Then, we show that knocking down cyd-1 in the uterine tissue of an IIS receptor mutant arrests oogenesis at the pachytene stage of meiosis-1 in a DAF-16-dependent manner. We observe activated DAF-16-dependent deterioration of the somatic gonadal tissues like the sheath cells, and transcriptional de-regulation of the sperm-to-oocyte switch genes which may be the underlying reason for the absence of oogenesis. Deleting DAF-16 releases the arrest and leads to restoration of the somatic gonad but poor-quality oocytes are produced. Together, our study reveals the unrecognized cell non-autonomous interaction of Cyclin D/CYD-1 and FOXO/DAF-16 in the regulation of oogenesis and reproductive fidelity.
Collapse
Affiliation(s)
- Umanshi Rautela
- Molecular Aging Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Gautam Chandra Sarkar
- Molecular Aging Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Ayushi Chaudhary
- Molecular Aging Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Debalina Chatterjee
- Molecular Aging Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Mohtashim Rosh
- Molecular Genetics Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | - Arnab Mukhopadhyay
- Molecular Aging Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
73
|
Chen S, Sun D, Zhang S, Xu L, Wang N, Li H, Xu X, Wei F. TIN2 modulates FOXO1 mitochondrial shuttling to enhance oxidative stress-induced apoptosis in retinal pigment epithelium under hyperglycemia. Cell Death Differ 2024; 31:1487-1505. [PMID: 39080375 PMCID: PMC11519896 DOI: 10.1038/s41418-024-01349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 10/30/2024] Open
Abstract
Progressive dysfunction of the retinal pigment epithelium (RPE) and the adjacent photoreceptor cells in the outer retina plays a pivotal role in the pathogenesis of diabetic retinopathy (DR). Here, we observed a marked increase in oxidative stress-induced apoptosis in parallel with higher expression of telomeric protein TIN2 in RPE cells under hyperglycemia in vivo and in vitro. Delving deeper, we confirm that high glucose-induced elevation of mitochondria-localized TIN2 compromises mitochondrial activity and weakens the intrinsic antioxidant defense, thereby leading to the activation of mitochondria-dependent apoptotic pathways. Mechanistically, mitochondrial TIN2 promotes the phosphorylation of FOXO1 and its relocation to the mitochondria. Such translocation of transcription factor FOXO1 not only promotes its binding to the D-loop region of mitochondrial DNA-resulting in the inhibition of mitochondrial respiration-but also hampers its availability to nuclear target DNA, thereby undermining the intrinsic antioxidant defense. Moreover, TIN2 knockdown effectively mitigates oxidative-induced apoptosis in diabetic mouse RPE by preserving mitochondrial homeostasis, which concurrently prevents secondary photoreceptor damage. Our study proposes the potential of TIN2 as a promising molecular target for therapeutic interventions for diabetic retinopathy, which emphasizes the potential significance of telomeric proteins in the regulation of metabolism and mitochondrial function. Created with BioRender ( https://www.biorender.com/ ).
Collapse
Affiliation(s)
- Shimei Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Dandan Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuchang Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Li Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Ning Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Huiming Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
74
|
Lee DH, Lee HJ, Yang G, Kim DY, Kim JU, Yook TH, Lee JH, Kim HJ. A novel treatment strategy targeting cellular pathways with natural products to alleviate sarcopenia. Phytother Res 2024; 38:5033-5051. [PMID: 39099170 DOI: 10.1002/ptr.8301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
Sarcopenia is a condition marked by a significant reduction in muscle mass and strength, primarily due to the aging process, which critically impacts muscle protein dynamics, metabolic functions, and overall physical functionality. This condition leads to increased body fat and reduced daily activity, contributing to severe health issues and a lower quality of life among the elderly. Recognized in the ICD-10-CM only in 2016, sarcopenia lacks definitive treatment options despite its growing prevalence and substantial social and economic implications. Given the aging global population, addressing sarcopenia has become increasingly relevant and necessary. The primary causes include aging, cachexia, diabetes, and nutritional deficiencies, leading to imbalances in protein synthesis and degradation, mitochondrial dysfunction, and hormonal changes. Exercise remains the most effective intervention, but it is often impractical for individuals with limited mobility, and pharmacological options such as anabolic steroids and myostatin inhibitors are not FDA-approved and are still under investigation. This review is crucial as it examines the potential of natural products as a novel treatment strategy for sarcopenia, targeting multiple mechanisms involved in its pathogenesis. By exploring natural products' multi-targeted effects, this study aims to provide innovative and practical solutions for sarcopenia management. Therefore, this review indicates significant improvements in muscle mass and function with the use of specific natural compounds, suggesting promising alternatives for those unable to engage in regular physical activity.
Collapse
Affiliation(s)
- Da Hee Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Hye Jin Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Gabsik Yang
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Dae Yong Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jong Uk Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Tae Han Yook
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| | - Jun Ho Lee
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
- Da Capo Co., Ltd., Jeonju-si, Republic of Korea
| | - Hong Jun Kim
- College of Korean Medicine, Woosuk University, Jeonju-si, Republic of Korea
| |
Collapse
|
75
|
Phuagkhaopong S, Janpattanapichai J, Sirirak N, Khemawoot P, Vivithanaporn P, Suknuntha K. Transcriptome analysis reveals a role of FOXO3 in antileukemia/lymphoma properties of panduratin A. Sci Rep 2024; 14:24795. [PMID: 39433897 PMCID: PMC11494127 DOI: 10.1038/s41598-024-75630-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Boesenbergia rotunda, commonly known as fingerroot, is a medicinal and culinary plant native to the Indochina Peninsula. We found that panduratin A (Pan-A), one of the compounds present in the rhizome extract of fingerroot, inhibited cell proliferation, induced apoptosis, and promoted cell cycle arrest at G0/G1 phase in multiple hematologic malignant cell lines including leukemia and lymphoma lines. Pan-A inhibited these activities in leukemia and lymphoma cells in a concentration-dependent manner. High-throughput transcriptome analysis indicated that Pan-A is involved in the cell cycle, cellular senescence, apoptosis, and multiple canonical signaling pathways in lymphoma cells. The Forkhead box O (FOXO) transcription factor family was identified as a potential target of Pan-A. Western blot showed elevated caspase 7 and cPARP/PARP in the B-cell lymphoma cells after treatment with Pan-A. The inhibitory effects were accompanied by stimulation of Akt signaling and phosphorylation of FOXO3. Immunohistochemistry of tissues from patients with B-cell lymphoma revealed detectable levels of FOXO3 protein specifically in neoplastic B cells. Overall, our results highlight FOXO3 as a player underlying antileukemia/lymphoma effects of Pan-A.
Collapse
Affiliation(s)
- Suttinee Phuagkhaopong
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jiranan Janpattanapichai
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Noppavut Sirirak
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Kran Suknuntha
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
76
|
Saini S, Tuli HS, Saini RV, Saini AK, Sak K, Kaur D, Shahwan M, Chauhan R, Chauhan A. Flavonoid-Mediated Suppression of Tumor Angiogenesis: Roles of Ang-Tie/PI3K/AKT. PATHOPHYSIOLOGY 2024; 31:596-607. [PMID: 39449525 PMCID: PMC11503374 DOI: 10.3390/pathophysiology31040043] [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: 08/23/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Angiogenesis is a process involved in the formation of new blood capillaries from pre-existing ones. It is regulated by several anti-angiogenic molecules involved in tumor growth and metastasis. The endothelial angiopoietin Ang-Tie/PI3K/AKT growth receptor pathway is necessary for healthy vascular development. The activation of AKT is controlled by a multistep process involving phosphoinositide 3-kinase (PI3K). This article aims to provide an overview of the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways and the potential of flavonoids as anti-angiogenic drugs. Flavonoids have shown great potential in preventing angiogenesis by targeting signaling pathways and exhibit additional anti-cancer properties. Research studies have revealed that the currently available anti-angiogenic drugs do not meet the safety and efficacy standards for treating tumor growth. Phytocompounds have long been a valuable resource for the development of novel therapeutic drugs. This article explores recent findings explaining the role and mechanism of the Ang-Tie/PI3K/AKT signaling pathways, as well as the interaction of flavonoids with angiogenic signaling pathways as a novel therapeutic approach. Several investigations have shown that synergistic studies of natural phytocompounds have great potential to target these pathways to inhibit tumor growth. Therefore, flavonoid-based medications may offer a more effective synergistic strategy to treat cancer.
Collapse
Affiliation(s)
- Shallu Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Reena V. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | - Adesh K. Saini
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala 133207, India; (R.V.S.); (A.K.S.)
| | | | - Damandeep Kaur
- University Centre for Research and Development, Chandigarh University, Mohali 140413, India;
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 4184, United Arab Emirates;
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to Be University, Dehradun 248002, India;
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Sector-125, Noida 201303, India;
| |
Collapse
|
77
|
Cao J, Tao C, Qin X, Wu K, Yang H, Liu C, Cheng T. PI3K-Akt-SGF1-Dimm pathway mediates the nutritional regulation of silk protein synthesis in Bombyx mori. Int J Biol Macromol 2024; 278:134650. [PMID: 39128739 DOI: 10.1016/j.ijbiomac.2024.134650] [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: 05/30/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The efficient synthesis of silk protein is heavily reliant on the ingestion of massive nutrients during the peak growth phase in the silkworm. However, the molecular mechanism of nutritional regulation of silk protein synthesis remains unknown. In this study, we investigated the impact of nutrient deficiency on the synthesis of silk protein. Nutritional deficiency led to a reduction in silk yield, accompanied by decreased levels of silk proteins and fibroin heavy chain (FibH)-activating transcription factors SGF1 and Dimm. Furthermore, insulin enhanced the protein levels of SGF1 and Dimm, which can be attenuated by specific inhibitors of PI3K. Co-immunoprecipitation analysis showed that the nutrient pathway factor protein kinase B (Akt) could interact with SGF1 protein. Knockdown of Akt reduced the phosphorylation level of SGF1 and impedes its nuclear translocation. Further studies revealed that SGF1 was directly bound to Fkh site in the 22-43 region upstream of ATG of Dimm gene to activate its transcription. In conclusion, during the peak growth phase, nutrition promotes the massive synthesis of silk protein through the PI3K-Akt-SGF1-Dimm pathway. This study offers valuable insights into the efficient synthesis of silk proteins and establishes a theoretical foundation for improving silk yield.
Collapse
Affiliation(s)
- Jun Cao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Cuicui Tao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Xiaodan Qin
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Keli Wu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Hongguo Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| | - Chun Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China.
| | - Tingcai Cheng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China.
| |
Collapse
|
78
|
Colino-Lage H, Guerrero-Gómez D, Gómez-Orte E, González X, Martina JA, Dansen TB, Ayuso C, Askjaer P, Puertollano R, Irazoqui JE, Cabello J, Miranda-Vizuete A. Regulation of Caenorhabditis elegans HLH-30 subcellular localization dynamics: Evidence for a redox-dependent mechanism. Free Radic Biol Med 2024; 223:369-383. [PMID: 39059513 PMCID: PMC11977398 DOI: 10.1016/j.freeradbiomed.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Basic Helix-Loop-Helix (bHLH) transcription factors TFEB/TFE3 and HLH-30 are key regulators of autophagy induction and lysosomal biogenesis in mammals and C. elegans, respectively. While much is known about the regulation of TFEB/TFE3, how HLH-30 subcellular dynamics and transactivation are modulated are yet poorly understood. Thus, elucidating the regulation of C. elegans HLH-30 will provide evolutionary insight into the mechanisms governing the function of bHLH transcription factor family. We report here that HLH-30 is retained in the cytoplasm mainly through its conserved Ser201 residue and that HLH-30 physically interacts with the 14-3-3 protein FTT-2 in this location. The FoxO transcription factor DAF-16 is not required for HLH-30 nuclear translocation upon stress, despite that both proteins partner to form a complex that coordinately regulates several organismal responses. Similar as described for DAF-16, the importin IMB-2 assists HLH-30 nuclear translocation, but constitutive HLH-30 nuclear localization is not sufficient to trigger its distinctive transcriptional response. Furthermore, we identify FTT-2 as the target of diethyl maleate (DEM), a GSH depletor that causes a transient nuclear translocation of HLH-30. Together, our work demonstrates that the regulation of TFEB/TFE3 and HLH-30 family members is evolutionarily conserved and that, in addition to a direct redox regulation through its conserved single cysteine residue, HLH-30 can also be indirectly regulated by a redox-dependent mechanism, probably through FTT-2 oxidation.
Collapse
Affiliation(s)
- Hildegard Colino-Lage
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - David Guerrero-Gómez
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Eva Gómez-Orte
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain
| | - Xavier González
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - José A Martina
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, CG Utrecht, the Netherlands
| | - Cristina Ayuso
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Junta de Andalucía, Seville, Spain
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Javier E Irazoqui
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Juan Cabello
- Centro de Investigación Biomédica de la Rioja (CIBIR), Logroño, La Rioja, Spain.
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.
| |
Collapse
|
79
|
Keshri PK, Singh SP. Unraveling the AKT/ERK cascade and its role in Parkinson disease. Arch Toxicol 2024; 98:3169-3190. [PMID: 39136731 DOI: 10.1007/s00204-024-03829-9] [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: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
Parkinson disease represents a significant and growing burden on global healthcare systems, necessitating a deeper understanding of their underlying molecular mechanisms for the development of effective treatments. The AKT and ERK pathways play crucial roles in the disease, influencing multiple cellular pathways that support neuronal survival. Researchers have made notable progress in uncovering how these pathways are controlled by upstream kinases and how their downstream effects contribute to cell signalling. However, as we delve deeper into their intricacies, we encounter increasing complexity, compounded by the convergence of multiple signalling pathways. Many of their targets overlap with those of other kinases, and they not only affect specific substrates but also influence entire signalling networks. This review explores the intricate interplay of the AKT/ERK pathways with several other signalling cascades, including oxidative stress, endoplasmic reticulum stress, calcium homeostasis, inflammation, and autophagy, in the context of Parkinson disease. We discuss how dysregulation of these pathways contributes to disease progression and neuronal dysfunction, highlighting potential therapeutic targets for intervention. By elucidating the complex network of interactions between the AKT/ERK pathways and other signalling cascades, this review aims to provide insights into the pathogenesis of Parkinson disease and describe the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
80
|
Liu W, Zhang Y, Nie Y, Liu Y, Li Z, Zhang Z, Gong B, Ma M. AGBL2 promotes renal cell carcinoma cells proliferation and migration via α-tubulin detyrosination. Heliyon 2024; 10:e37086. [PMID: 39315218 PMCID: PMC11417249 DOI: 10.1016/j.heliyon.2024.e37086] [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: 02/17/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background AGBL2's role in tumorigenesis and cancer progression has been reported in several cancer studies, and it is closely associated with α-tubulin detyrosination. The roles of AGBL2 and α-tubulin detyrosination in renal cell carcinoma (RCC) pathogenesis remain unclear and require further investigation. Methods In this study, we conducted an analysis of AGBL2 expression differences between renal clear cell carcinoma tissues and normal tissues using data from The Cancer Genome Atlas (TCGA). We performed a comprehensive prognostic analysis of AGBL2 in Kidney Renal Clear Cell Carcinoma (KIRC) using univariate and multivariate Cox regression. Based on the results of the Cox analysis, we constructed a prognostic model to assess its predictive capabilities. Receiver Operating Characteristic (ROC) analysis confirmed the diagnostic value of AGBL2 in renal cancer. We conducted further validation by analyzing cancer tissue samples and renal cancer cell lines, which confirmed the role of AGBL2 in promoting RCC cell proliferation and migration through in vitro experiments. Additionally, we verified the impact of AGBL2's detyrosination on α-tubulin using the tubulin carboxypeptidase (TCP) inhibitor parthenolide. Finally, we performed sequencing analysis on AGBL2 knockdown 786-O cells to investigate the correlation between AGBL2, immune infiltration, and AKT phosphorylation. Moreover, we experimentally demonstrated the enhancing effect of AGBL2 on AKT phosphorylation. Results TCGA analysis revealed a significant increase in AGBL2 expression in RCC patients, which was correlated with poorer overall survival (OS), disease-specific survival (DSS), and progression-free intervals (PFI). According to the analysis results, we constructed column-line plots to predict the 1-, 3-, and 5-year survival outcomes in RCC patients. Additionally, the calibration plots assessing the model's performance exhibited favorable agreement with the predicted outcomes. And the ROC curves showed that AGBL2 showed good diagnostic performance in KIRC (AUC = 0.836)). Cell phenotyping assays revealed that AGBL2 knockdown in RCC cells significantly inhibited cell proliferation and migration. Conversely, overexpression of AGBL2 resulted in increased cell proliferation and migration in RCC cells. We observed that AGBL2 is predominantly located in the nucleus and can elevate the detyrosination level of α-tubulin in RCC cells. Moreover, the enhancement of RCC cell proliferation and migration by AGBL2 was partially inhibited after treatment with the TCP inhibitor parthenolide. Analysis of the sequencing data revealed that AGBL2 is associated with a diverse array of biological processes, encompassing signal transduction and immune infiltration. Interestingly, AGBL2 expression exhibited a negative correlation with the majority of immune cell infiltrations. Additionally, AGBL2 was found to enhance the phosphorylation of AKT in RCC cells. Conclusion Our study suggests that AGBL2 fosters RCC cell proliferation and migration by enhancing α-tubulin detyrosination. Moreover, elevated AGBL2 expression increases phosphorylation of AKT in RCC cells.
Collapse
Affiliation(s)
- Wei Liu
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yifei Zhang
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yechen Nie
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yifu Liu
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhongqi Li
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhicheng Zhang
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Binbin Gong
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ming Ma
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Department of Urology, Gaoxin Branch of The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, China
| |
Collapse
|
81
|
Yi Y, Liu G, Li Y, Wang C, Zhang B, Lou H, Yu S. Baicalin Ameliorates Depression-like Behaviors via Inhibiting Neuroinflammation and Apoptosis in Mice. Int J Mol Sci 2024; 25:10259. [PMID: 39408591 PMCID: PMC11476789 DOI: 10.3390/ijms251910259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/09/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Depression is a common neuropsychiatric disease which brings an increasing burden to all countries globally. Baicalin, a flavonoid extracted from the dried roots of Scutellaria, has been reported to exert anti-inflammatory, antioxidant, and neuroprotective effects in the treatment of depression. However, the potential biological mechanisms underlying its antidepressant effect are still unclear. In the present study, we conducted extensive research on the potential mechanisms of baicalin's antidepressant effect using the methods of network pharmacology, including overlapped terms-based analysis, protein-protein interaction (PPI) network topology analysis, and enrichment analysis. Moreover, these results were further verified through molecular docking, weighted gene co-expression network analysis (WGCNA), differential gene expression analysis, and subsequent animal experiments. We identified forty-one genes as the targets of baicalin in the treatment of depression, among which AKT1, IL6, TP53, IL1B, and CASP3 have higher centrality in the more core position. Meanwhile, the roles of peripheral genes derived from direct potential targets were also observed. Our study suggested that biological processes, such as inflammatory reaction, apoptosis, and oxidative stress, may be involved in the therapeutic process of baicalin on depression. These mechanisms were validated at the level of structure, gene, protein, and signaling pathway in the present study. Taken together, these findings propose a new perspective on the potential mechanisms underlying baicalin's antidepressant effect, and also provide a new basis and clarified perspective for its clinical application.
Collapse
Affiliation(s)
- Yuhang Yi
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Y.Y.); (G.L.); (Y.L.); (C.W.)
| | - Guiyu Liu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Y.Y.); (G.L.); (Y.L.); (C.W.)
| | - Ye Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Y.Y.); (G.L.); (Y.L.); (C.W.)
| | - Changmin Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Y.Y.); (G.L.); (Y.L.); (C.W.)
| | - Bin Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (B.Z.); (H.L.)
| | - Haiyan Lou
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (B.Z.); (H.L.)
| | - Shuyan Yu
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; (Y.Y.); (G.L.); (Y.L.); (C.W.)
- Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, Jinan 250012, China
| |
Collapse
|
82
|
Niimi K, Nakae J, Kubota Y, Inagaki S, Furuyama T. Macrophages play a crucial role in vascular smooth muscle cell coverage. Development 2024; 151:dev203080. [PMID: 39166965 DOI: 10.1242/dev.203080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/11/2024] [Indexed: 08/23/2024]
Abstract
The microvascular system consists of two cell types: endothelial and mural (pericytes and vascular smooth muscle cells; VSMCs) cells. Communication between endothelial and mural cells plays a pivotal role in the maintenance of vascular homeostasis; however, in vivo molecular and cellular mechanisms underlying mural cell development remain unclear. In this study, we found that macrophages played a crucial role in TGFβ-dependent pericyte-to-VSMC differentiation during retinal vasculature development. In mice with constitutively active Foxo1 overexpression, substantial accumulation of TGFβ1-producing macrophages and pericytes around the angiogenic front region was observed. Additionally, the TGFβ-SMAD pathway was activated in pericytes adjacent to macrophages, resulting in excess ectopic α-smooth muscle actin-positive VSMCs. Furthermore, we identified endothelial SEMA3C as an attractant for macrophages. In vivo neutralization of SEMA3C rescued macrophage accumulation and ectopic VSMC phenotypes in the mice, as well as drug-induced macrophage depletion. Therefore, macrophages play an important physiological role in VSMC development via the FOXO1-SEMA3C pathway.
Collapse
Affiliation(s)
- Kenta Niimi
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Hara 281-1, Mure, Takamatsu, Kagawa 761-0123, Japan
| | - Jun Nakae
- Department of Physiology, International University of Health and Welfare School of Medicine, 4-3 Kozu-no-Mori, Narita 286-8686, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35-Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shinobu Inagaki
- Department of Physical Therapy, Osaka Yukioka College of Health Science, Sojiji 1-1-41, Ibaraki, Osaka 567-0801, Japan
| | - Tatsuo Furuyama
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Hara 281-1, Mure, Takamatsu, Kagawa 761-0123, Japan
| |
Collapse
|
83
|
Park SH. Role of Phytochemicals in Treatment of Aging and Cancer: Focus on Mechanism of FOXO3 Activation. Antioxidants (Basel) 2024; 13:1099. [PMID: 39334758 PMCID: PMC11428386 DOI: 10.3390/antiox13091099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
There have been many studies reporting that the regular consumption of fruits and vegetables is associated with reduced risks of cancer and age-related chronic diseases. Recent studies have demonstrated that reducing reactive oxygen species and inflammation by phytochemicals derived from natural sources can extend lifespans in a range of model organisms. Phytochemicals derived from fruits and vegetables have been known to display both preventative and suppressive activities against various types of cancer via in vitro and in vivo research by interfering with cellular processes critical for tumor development. The current challenge lies in creating tailored supplements containing specific phytochemicals for individual needs. Achieving this goal requires a deeper understanding of the molecular mechanisms through which phytochemicals affect human health. In this review, we examine recently (from 2010 to 2024) reported plant extracts and phytochemicals with established anti-aging and anti-cancer effects via the activation of FOXO3 transcriptional factor. Additionally, we provide an overview of the cellular and molecular mechanisms by which these molecules exert their anti-aging and anti-cancer effects in specific model systems. Lastly, we discuss the limitations of the current research approach and outline for potential future directions in this field.
Collapse
Affiliation(s)
- See-Hyoung Park
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
84
|
Augustin HG, Koh GY. A systems view of the vascular endothelium in health and disease. Cell 2024; 187:4833-4858. [PMID: 39241746 DOI: 10.1016/j.cell.2024.07.012] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 09/09/2024]
Abstract
The dysfunction of blood-vessel-lining endothelial cells is a major cause of mortality. Although endothelial cells, being present in all organs as a single-cell layer, are often conceived as a rather inert cell population, the vascular endothelium as a whole should be considered a highly dynamic and interactive systemically disseminated organ. We present here a holistic view of the field of vascular research and review the diverse functions of blood-vessel-lining endothelial cells during the life cycle of the vasculature, namely responsive and relaying functions of the vascular endothelium and the responsive roles as instructive gatekeepers of organ function. Emerging translational perspectives in regenerative medicine, preventive medicine, and aging research are developed. Collectively, this review is aimed at promoting disciplinary coherence in the field of angioscience for a broader appreciation of the importance of the vasculature for organ function, systemic health, and healthy aging.
Collapse
Affiliation(s)
- Hellmut G Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Division of Vascular Oncology and Metastasis, German Cancer Research Center Heidelberg (DKFZ), 69120 Heidelberg, Germany.
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
85
|
Abstract
Behavioral plasticity allows animals to modulate their behavior based on experience and environmental conditions. Caenorhabditis elegans exhibits experience-dependent changes in its behavioral responses to various modalities of sensory cues, including odorants, salts, temperature, and mechanical stimulations. Most of these forms of behavioral plasticity, such as adaptation, habituation, associative learning, and imprinting, are shared with other animals. The C. elegans nervous system is considerably tractable for experimental studies-its function can be characterized and manipulated with molecular genetic methods, its activity can be visualized and analyzed with imaging approaches, and the connectivity of its relatively small number of neurons are well described. Therefore, C. elegans provides an opportunity to study molecular, neuronal, and circuit mechanisms underlying behavioral plasticity that are either conserved in other animals or unique to this species. These findings reveal insights into how the nervous system interacts with the environmental cues to generate behavioral changes with adaptive values.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
- Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yuichi Iino
- Department of Biological Sciences, University of Tokyo, Tokyo 113-0032, Japan
| | - William R Schafer
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, Cambridgeshire CB2 0QH, UK
- Department of Biology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
86
|
Bao W, Lyu J, Feng G, Guo L, Zhao D, You K, Liu Y, Li H, Du P, Chen D, Shen X. Aloe emodin promotes mucosal healing by modifying the differentiation fate of enteroendocrine cells via regulating cellular free fatty acid sensitivity. Acta Pharm Sin B 2024; 14:3964-3982. [PMID: 39309505 PMCID: PMC11413701 DOI: 10.1016/j.apsb.2024.05.027] [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: 12/25/2023] [Revised: 03/08/2024] [Accepted: 04/12/2024] [Indexed: 09/25/2024] Open
Abstract
The proper differentiation and reorganization of the intestinal epithelial cell population is critical to mucosal regeneration post injury. Label retaining cells (LRCs) expressing SRY-box transcription factor 9 (SOX9) promote epithelial repair by replenishing LGR5+ intestinal stem cells (ISCs). While, LRCs are also considered precursor cells for enteroendocrine cells (EECs) which exacerbate mucosal damage in inflammatory bowel disease (IBD). The factors that determine LRC-EEC differentiation and the effect of intervening in LRC-EEC differentiation on IBD remain unclear. In this study, we investigated the effects of a natural anthraquinone called aloe emodin (derived from the Chinese herb rhubarb) on mucosal healing in IBD models. Our findings demonstrated that aloe emodin effectively interfered with the differentiation to EECs and preserved a higher number of SOX9+ LRCs, thereby promoting mucosal healing. Furthermore, we discovered that aloe emodin acted as an antagonist of free fatty acid receptors (FFAR1), suppressing the FFAR1-mediated Gβγ/serine/threonine-protein kinase (AKT) pathway and promoting the translocation of forkhead box protein O1 (FOXO1) into the nucleus, ultimately resulting in the intervention of differentiation fate. These findings reveal the effect of free fatty acid accessibility on EEC differentiation and introduce a strategy for promoting mucosal healing in IBD by regulating the FFAR1/AKT/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Weilian Bao
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Jiaren Lyu
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Guize Feng
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Linfeng Guo
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Dian Zhao
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Keyuan You
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Yang Liu
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Haidong Li
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Peng Du
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Shanghai Colorectal Cancer Research Center, Shanghai 200092, China
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201210, China
| | - Xiaoyan Shen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201210, China
| |
Collapse
|
87
|
Kim JH, Choi MS, Auger C, Lee KW, Schini-Kerth VB. Polyphenol-rich Aronia melanocarpa juice sustains eNOS activation through phosphorylation and expression via redox-sensitive pathways in endothelial cells. Food Sci Biotechnol 2024; 33:2865-2875. [PMID: 39184991 PMCID: PMC11339019 DOI: 10.1007/s10068-024-01546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 08/27/2024] Open
Abstract
A sustained formation of nitric oxide (NO) by endothelial nitric oxide synthase (eNOS) is crucial to safeguard the vascular system against the development of cardiovascular diseases. This study investigated the prolonged phosphorylation and expression of eNOS induced by polyphenol-rich Aronia melanocarpa juice (AMJ), along with its underlying mechanisms. The findings revealed that AMJ triggered concentration- and time-dependent increases in eNOS phosphorylation and expression, leading to sustained NO production for 15 h. Investigations with various enzymes and inhibitors revealed that the effect of AMJ was associated with redox sensitivity, activating the PI3-kinase/Akt, JNK, and p38 MAPK pathways. These pathways led to the inactivation of transcription factors FoxO1 and FoxO3a through phosphorylation, relieving their repression on eNOS expression. Therefore, the capability of AMJ to consistently trigger prolonged eNOS phosphorylation and expression via complex redox-sensitive pathways highlights its potential for maintaining vascular health and preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Jong Hun Kim
- Department of Food Science and Biotechnology and Institute for Basic Sciences, Sungshin Women’s University, Seoul, 01133 Republic of Korea
| | - Min Sik Choi
- Lab of Pharmacology, College of Pharmacy, Dongduk Women’s University, Seoul, 02748 Republic of Korea
| | - Cyril Auger
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, Strasbourg, France
| | - Ki Won Lee
- Department of Agricultural Biotechnology, Biomodulation Major, Seoul National University, Seoul, 08826 Republic of Korea
- Bio-MAX Institute, Seoul National University, Seoul, 08826 Republic of Korea
- Advanced Institute of Convergence Technology, Seoul National University, Suwon, 16229 Republic of Korea
| | | |
Collapse
|
88
|
Al-Rawi DH, Lettera E, Li J, DiBona M, Bakhoum SF. Targeting chromosomal instability in patients with cancer. Nat Rev Clin Oncol 2024; 21:645-659. [PMID: 38992122 DOI: 10.1038/s41571-024-00923-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2024] [Indexed: 07/13/2024]
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and a driver of metastatic dissemination, therapeutic resistance, and immune evasion. CIN is present in 60-80% of human cancers and poses a formidable therapeutic challenge as evidenced by the lack of clinically approved drugs that directly target CIN. This limitation in part reflects a lack of well-defined druggable targets as well as a dearth of tractable biomarkers enabling direct assessment and quantification of CIN in patients with cancer. Over the past decade, however, our understanding of the cellular mechanisms and consequences of CIN has greatly expanded, revealing novel therapeutic strategies for the treatment of chromosomally unstable tumours as well as new methods of assessing the dynamic nature of chromosome segregation errors that define CIN. In this Review, we describe advances that have shaped our understanding of CIN from a translational perspective, highlighting both challenges and opportunities in the development of therapeutic interventions for patients with chromosomally unstable cancers.
Collapse
Affiliation(s)
- Duaa H Al-Rawi
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emanuele Lettera
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody DiBona
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
89
|
Forte G, Donghia R, Lepore Signorile M, Tatoli R, Bonfiglio C, Losito F, De Marco K, Manghisi A, Guglielmi FA, Disciglio V, Fasano C, Sanese P, Cariola F, Buonadonna AL, Grossi V, Giannelli G, Simone C. Exploring the Relationship of rs2802292 with Diabetes and NAFLD in a Southern Italian Cohort-Nutrihep Study. Int J Mol Sci 2024; 25:9512. [PMID: 39273459 PMCID: PMC11394752 DOI: 10.3390/ijms25179512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
Background: The minor G-allele of FOXO3 rs2802292 is associated with human longevity. The aim of this study was to test the protective effect of the variant against the association with type 2 Diabetes and NAFLD. Methods: rs2802292 was genotyped in a large population of middle-aged subjects (n = 650) from a small city in Southern Italy. All participants were interviewed to collect information about lifestyle and dietary habits; clinical characteristics were recorded, and blood samples were collected from all subjects. The association between rs2802292 and NAFLD or diabetes was tested using a logistic model and mediation analysis adjusted for covariates. Results: Overall, the results indicated a statistical association between diabetes and rs2802292, especially for the TT genotype (OR = 2.14, 1.01 to 4.53 95% C.I., p = 0.05) or in any case for those who possess the G-allele (OR = 0.45, 0.25 to 0.81 95% C.I., p = 0.008). Furthermore, we found a mediation effect of rs2802292 on diabetes (as mediator) and NAFLD. There is no direct relationship between rs2802292 and NAFLD, but the effect is direct (β = 0.10, -0.003 to 0.12 95% C.I., p = 0.04) on diabetes, but only in TT genotypes. Conclusions: The data on our cohort indicate that the longevity-associated FOXO3 variant may have protective effects against diabetes and NAFLD.
Collapse
Affiliation(s)
- Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Rossella Donghia
- Data Science Unit, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (R.D.); (R.T.); (C.B.)
| | - Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Rossella Tatoli
- Data Science Unit, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (R.D.); (R.T.); (C.B.)
| | - Caterina Bonfiglio
- Data Science Unit, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (R.D.); (R.T.); (C.B.)
| | - Francesco Losito
- Gastroenterology Unit, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Katia De Marco
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Andrea Manghisi
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Filomena Anna Guglielmi
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Vittoria Disciglio
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Paola Sanese
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Filomena Cariola
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Antonia Lucia Buonadonna
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology, IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (M.L.S.); (K.D.M.); (A.M.); (F.A.G.); (V.D.); (C.F.); (P.S.); (F.C.); (A.L.B.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
90
|
Xiao Y, Deng F, Luo Y, Wang T. Pharmaceutical inhibition of BCL6 ameliorates resistance to imatinib in chronic myeloid leukemia. Heliyon 2024; 10:e36640. [PMID: 39258188 PMCID: PMC11386027 DOI: 10.1016/j.heliyon.2024.e36640] [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: 10/25/2023] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
The tyrosine kinase inhibitors (TKIs) have improved overall survival of CML (chronic myeloid leukemia) patients and allow them to experience normal life expectancy. However, relapse and drug resistance remain the main challenges in the clinical treatment of CML. The B-cell lymphoma 6 (BCL6) is essential to regulation of multiple function such as immune response and lymphomagenesis in lymph node germinal cells. Recent studies have shown that BCL6 is required for the maintenance of leukemia stem cells in CML, but the expression of Bcl-6 in response to Imatinib and the underlying mechanism are still unclear. Here, we found that BCL6 is expressed at high levels in primary CML bone marrow samples and CML TKI-resistance cell lines. CML cells with higher levels of BCL6 were generally sensitive to treatment with BCL6 inhibitors, BI-3812. Treatment of CML cells with BCL6 inhibitor and TKIs suggested enhanced anti-leukemia activity. In summary, our findings suggest BCL6 as a therapeutic target for the treatment of CML.
Collapse
Affiliation(s)
- Yingying Xiao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Deng
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Luo
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Teng Wang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
91
|
Wang M, Dai B, Liu Q, Zhang X. Prognostic and immunological implications of heterogeneous cell death patterns in prostate cancer. Cancer Cell Int 2024; 24:297. [PMID: 39182081 PMCID: PMC11344416 DOI: 10.1186/s12935-024-03462-7] [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: 02/05/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Prostate cancer is one of the most common cancers in men with a significant proportion of patients developing biochemical recurrence (BCR) after treatment. Programmed cell death (PCD) mechanisms are known to play critical roles in tumor progression and can potentially serve as prognostic and therapeutic biomarkers in PCa. This study aimed to develop a prognostic signature for BCR in PCa using PCD-related genes. MATERIALS AND METHODS We conducted an analysis of 19 different modes of PCD to develop a comprehensive model. Bulk transcriptomic, single-cell transcriptomic, genomic, and clinical data were collected from multiple cohorts, including TCGA-PRAD, GSE58812, METABRIC, GSE21653, and GSE193337. We analyzed the expression and mutations of the 19 PCD modes and constructed, evaluated, and validated the model. RESULTS Ten PCD modes were found to be associated with BCR in PCa, with specific PCD patterns exhibited by various cell components within the tumor microenvironment. Through Lasso Cox regression analysis, we established a Programmed Cell Death Index (PCDI) utilizing an 11-gene signature. High PCDI values were validated in five independent datasets and were found to be associated with an increased risk of BCR in PCa patients. Notably, older age and advanced T and N staging were associated with higher PCDI values. By combining PCDI with T staging, we constructed a nomogram with enhanced predictive performance. Additionally, high PCDI values were significantly correlated with decreased drug sensitivity, including drugs such as Docetaxel and Methotrexate. Patients with lower PCDI values demonstrated higher immunophenoscores (IPS), suggesting a potentially higher response rate to immune therapy. Furthermore, PCDI was associated with immune checkpoint genes and key components of the tumor microenvironment, including macrophages, T cells, and NK cells. Finally, clinical specimens validated the differential expression of PCDI-related PCDRGs at both the gene and protein levels. CONCLUSION In conclusion, we developed a novel PCD-based prognostic feature that successfully predicted BCR in PCa patients and provided insights into drug sensitivity and potential response to immune therapy. These findings have significant clinical implications for the treatment of PCa.
Collapse
Affiliation(s)
- Ming Wang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, China
| | - Bangshun Dai
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, China
| | - Qiushi Liu
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, China
| | - Xiansheng Zhang
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, China.
| |
Collapse
|
92
|
Lyu Y, Meng Z, Hu Y, Jiang B, Yang J, Chen Y, Zhou J, Li M, Wang H. Mechanisms of mitophagy and oxidative stress in cerebral ischemia-reperfusion, vascular dementia, and Alzheimer's disease. Front Mol Neurosci 2024; 17:1394932. [PMID: 39169952 PMCID: PMC11335644 DOI: 10.3389/fnmol.2024.1394932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Neurological diseases have consistently represented a significant challenge in both clinical treatment and scientific research. As research has progressed, the significance of mitochondria in the pathogenesis and progression of neurological diseases has become increasingly prominent. Mitochondria serve not only as a source of energy, but also as regulators of cellular growth and death. Both oxidative stress and mitophagy are intimately associated with mitochondria, and there is mounting evidence that mitophagy and oxidative stress exert a pivotal regulatory influence on the pathogenesis of neurological diseases. In recent years, there has been a notable rise in the prevalence of cerebral ischemia/reperfusion injury (CI/RI), vascular dementia (VaD), and Alzheimer's disease (AD), which collectively represent a significant public health concern. Reduced levels of mitophagy have been observed in CI/RI, VaD and AD. The improvement of associated pathology has been demonstrated through the increase of mitophagy levels. CI/RI results in cerebral tissue ischemia and hypoxia, which causes oxidative stress, disruption of the blood-brain barrier (BBB) and damage to the cerebral vasculature. The BBB disruption and cerebral vascular injury may induce or exacerbate VaD to some extent. In addition, inadequate cerebral perfusion due to vascular injury or altered function may exacerbate the accumulation of amyloid β (Aβ) thereby contributing to or exacerbating AD pathology. Intravenous tissue plasminogen activator (tPA; alteplase) and endovascular thrombectomy are effective treatments for stroke. However, there is a narrow window of opportunity for the administration of tPA and thrombectomy, which results in a markedly elevated incidence of disability among patients with CI/RI. It is regrettable that there are currently no there are still no specific drugs for VaD and AD. Despite the availability of the U.S. Food and Drug Administration (FDA)-approved clinical first-line drugs for AD, including memantine, donepezil hydrochloride, and galantamine, these agents do not fundamentally block the pathological process of AD. In this paper, we undertake a review of the mechanisms of mitophagy and oxidative stress in neurological disorders, a summary of the clinical trials conducted in recent years, and a proposal for a new strategy for targeted treatment of neurological disorders based on both mitophagy and oxidative stress.
Collapse
Affiliation(s)
- Yujie Lyu
- Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, China
- Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| | - Zhipeng Meng
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yunyun Hu
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Bing Jiang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Jiao Yang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yiqin Chen
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Jun Zhou
- Xichang Hospital of Traditional Chinese Medicine, Xichang, China
| | - Mingcheng Li
- Qujing 69 Hospital, China RongTong Medical Healthcare Group Co. Ltd, Qujing, China
| | - Huping Wang
- Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Traditional Chinese Herbs and Prescription Innovation and Transformation of Gansu Province, Lanzhou, China
- Laboratory for TCM New Products Development Engineering of Gansu Province, Lanzhou, China
| |
Collapse
|
93
|
Cai X, Cao J, Wang L, Zou J, Li R, Sun P, Ding X, Zhang B, Liu Z, Pei X, Yang J, Zhan Y, Liu N, Liu T, Liang R, Gao J, Wang S. Liraglutide Protects Pancreatic Islet From Ischemic Injury by Reducing Oxidative Stress and Activating Akt Signaling During Cold Preservation to Improve Islet Transplantation Outcomes. Transplantation 2024; 108:e156-e169. [PMID: 38578708 DOI: 10.1097/tp.0000000000004949] [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] [Indexed: 04/07/2024]
Abstract
BACKGROUND Islet transplantation is a promising therapy for patients with type 1 diabetes. However, ischemic injury to the donor islets during cold preservation leads to reduced islet quality and compromises transplant outcome. Several studies imply that liraglutide, a glucagon-like peptide-1 receptor agonist, has a positive effect on promoting islet survival, but its impact on islet cold-ischemic injury remains unexplored. Therefore, the aim of this study was to investigate whether liraglutide can improve islet transplantation efficacy by inhibiting cold-ischemic injury and to explore the underlying mechanisms. METHODS Liraglutide was applied in a mouse pancreas preservation model and a human islets cold-preservation model, and islet viability, function, oxidative stress levels were evaluated. Furthermore, islet transplantation was performed in a syngeneic mouse model and a human-to-nude mouse islet xenotransplantation model. RESULTS The supplementation of liraglutide in preservation solution improved islet viability, function, and reduced cell apoptosis. Liraglutide inhibited the oxidative stress of cold-preserved pancreas or islets through upregulating the antioxidant enzyme glutathione levels, inhibiting reactive oxygen species accumulation, and maintaining the mitochondrial membrane integrity, which is associated with the activation of Akt signaling. Furthermore, the addition of liraglutide during cold preservation of donor pancreas or donor islets significantly improved the subsequent transplant outcomes in both syngeneic mouse islet transplantation model and human-to-nude mouse islet xenotransplantation model. CONCLUSIONS Liraglutide protects islets from cold ischemia-related oxidative stress during preservation and hence improved islet transplantation outcomes, and this protective effect of liraglutide in islets is associated with the activation of Akt signaling.
Collapse
Affiliation(s)
- Xiangheng Cai
- School of Medicine, Nankai University, Tianjin, China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jinglin Cao
- Department of Hepatobiliary Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Le Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jiaqi Zou
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Rui Li
- Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Peng Sun
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xuejie Ding
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Boya Zhang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zewen Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Xirui Pei
- First Clinical Department, The First Hospital of China Medical University, China Medical University, Shenyang, China
| | - Jiuxia Yang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yixiang Zhan
- School of Medicine, Nankai University, Tianjin, China
| | - Na Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Tengli Liu
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin, China
| | - Shusen Wang
- School of Medicine, Nankai University, Tianjin, China
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| |
Collapse
|
94
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
95
|
Rudnitsky E, Braiman A, Wolfson M, Muradian KK, Gorbunova V, Turgeman G, Fraifeld VE. Stem cell-derived extracellular vesicles as senotherapeutics. Ageing Res Rev 2024; 99:102391. [PMID: 38914266 DOI: 10.1016/j.arr.2024.102391] [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: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Cellular senescence (CS) is recognized as one of the hallmarks of aging, and an important player in a variety of age-related pathologies. Accumulation of senescent cells can promote a pro-inflammatory and pro-cancerogenic microenvironment. Among potential senotherapeutics are extracellular vesicles (EVs) (40-1000 nm), including exosomes (40-150 nm), that play an important role in cell-cell communications. Here, we review the most recent studies on the impact of EVs derived from stem cells (MSCs, ESCs, iPSCs) as well as non-stem cells of various types on CS and discuss potential mechanisms responsible for the senotherapeutic effects of EVs. The analysis revealed that (i) EVs derived from stem cells, pluripotent (ESCs, iPSCs) or multipotent (MSCs of various origin), can mitigate the cellular senescence phenotype both in vitro and in vivo; (ii) this effect is presumably senomorphic; (iii) EVs display cross-species activity, without apparent immunogenic responses. In summary, stem cell-derived EVs appear to be promising senotherapeutics, with a feasible application in humans.
Collapse
Affiliation(s)
- Ekaterina Rudnitsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Marina Wolfson
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Khachik K Muradian
- Department of Biology of Aging and Experimental Life Span Extension, State Institute of Gerontology of National Academy of Medical Sciences of Ukraine, Kiev 4114, Ukraine
| | - Vera Gorbunova
- Department of Biology, Rochester Aging Research Center, University of Rochester, Rochester, NY 14627, USA
| | - Gadi Turgeman
- Department of Molecular Biology, Faculty of Natural Sciences and Medical School, Ariel University, Ariel 40700, Israel.
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
96
|
Lazovic B, Nguyen HT, Ansarizadeh M, Wigge L, Kohl F, Li S, Carracedo M, Kettunen J, Krimpenfort L, Elgendy R, Richter K, De Silva L, Bilican B, Singh P, Saxena P, Jakobsson L, Hong X, Eklund L, Hicks R. Human iPSC and CRISPR targeted gene knock-in strategy for studying the somatic TIE2 L914F mutation in endothelial cells. Angiogenesis 2024; 27:523-542. [PMID: 38771392 PMCID: PMC11303492 DOI: 10.1007/s10456-024-09925-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/22/2024] [Indexed: 05/22/2024]
Abstract
Induced pluripotent stem cell (iPSC) derived endothelial cells (iECs) have emerged as a promising tool for studying vascular biology and providing a platform for modelling various vascular diseases, including those with genetic origins. Currently, primary ECs are the main source for disease modelling in this field. However, they are difficult to edit and have a limited lifespan. To study the effects of targeted mutations on an endogenous level, we generated and characterized an iPSC derived model for venous malformations (VMs). CRISPR-Cas9 technology was used to generate a novel human iPSC line with an amino acid substitution L914F in the TIE2 receptor, known to cause VMs. This enabled us to study the differential effects of VM causative mutations in iECs in multiple in vitro models and assess their ability to form vessels in vivo. The analysis of TIE2 expression levels in TIE2L914F iECs showed a significantly lower expression of TIE2 on mRNA and protein level, which has not been observed before due to a lack of models with endogenous edited TIE2L914F and sparse patient data. Interestingly, the TIE2 pathway was still significantly upregulated and TIE2 showed high levels of phosphorylation. TIE2L914F iECs exhibited dysregulated angiogenesis markers and upregulated migration capability, while proliferation was not affected. Under shear stress TIE2L914F iECs showed reduced alignment in the flow direction and a larger cell area than TIE2WT iECs. In summary, we developed a novel TIE2L914F iPSC-derived iEC model and characterized it in multiple in vitro models. The model can be used in future work for drug screening for novel treatments for VMs.
Collapse
Affiliation(s)
- Bojana Lazovic
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Hoang-Tuan Nguyen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Finnadvance Ltd., Oulu, Finland
| | - Mohammadhassan Ansarizadeh
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Leif Wigge
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Franziska Kohl
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Songyuan Li
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Miguel Carracedo
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Luc Krimpenfort
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ramy Elgendy
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kati Richter
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Laknee De Silva
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Bilada Bilican
- Translational Genomics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Pratik Saxena
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Xuechong Hong
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK.
| |
Collapse
|
97
|
Guo N, Wang X, Xu M, Bai J, Yu H, Le Zhang. PI3K/AKT signaling pathway: Molecular mechanisms and therapeutic potential in depression. Pharmacol Res 2024; 206:107300. [PMID: 38992850 DOI: 10.1016/j.phrs.2024.107300] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Depression is a serious global mental disorder. Numerous studies have found that depression may be closely related to decreased neurogenesis, neuroinflammation, neurotransmitter imbalance, and synaptic plasticity dysfunction. The pathogenesis of depression is complex and involves multiple signal transduction pathways and molecular changes. The PI3K/AKT pathway is an essential signaling pathways in neurons, which is widely expressed in emotion-related regions of the brain. Therefore, the PI3K/AKT pathway may play a moderating role in mood disorders. However, the role and mechanism of the PI3K/AKT signaling pathway in depression have not been fully described. This review systematically summarized the role of the PI3K/AKT signaling pathway in the pathogenesis of depression and discussed its potential in the treatment of depression. This will help in the treatment of depression and the development of antidepressants.
Collapse
Affiliation(s)
- Ningning Guo
- School of Mental Health, Jining Medical University, Jining, China
| | - Xin Wang
- Department of Radiation Therapy, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Muran Xu
- Clinical College, Jining Medical University, Jining, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, China.
| | - Hao Yu
- School of Mental Health, Jining Medical University, Jining, China.
| | - Le Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
98
|
Hu Y, Yi L, Yang Y, Wu Z, Kong M, Kang Z, Yang Z. Acetylation of FOXO1 activates Bim expression involved in CVB3 induced cardiomyocyte apoptosis. Apoptosis 2024; 29:1271-1287. [PMID: 38127284 PMCID: PMC11263423 DOI: 10.1007/s10495-023-01924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
Viral myocarditis (VMC) is the major reason for sudden cardiac death among both children and young adults. Of these, coxsackievirus B3 (CVB3) is the most common causative agent of myocarditis. Recently, the role of signaling pathways in the pathogenesis of VMC has been evaluated in several studies, which has provided a new perspective on identifying potential therapeutic targets for this hitherto incurable disease. In the present study, in vivo and in vitro experiments showed that CVB3 infection leads to increased Bim expression and triggers apoptosis. In addition, by knocking down Bim using RNAi, we further confirmed the biological function of Bim in apoptosis induced by CVB3 infection. We additionally found that Bim and forkhead box O1 class (FOXO1) inhibition significantly increased the viability of CVB3-infected cells while blocking viral replication and viral release. Moreover, CVB3-induced Bim expression was directly dependent on FOXO1 acetylation, which is catalyzed by the co-regulation of CBP and SirTs. Furthermore, the acetylation of FOXO1 was an important step in Bim activation and apoptosis induced by CVB3 infection. The findings of this study suggest that CVB3 infection induces apoptosis through the FOXO1 acetylation-Bim pathway, thus providing new insights for developing potential therapeutic targets for enteroviral myocarditis.
Collapse
Affiliation(s)
- Yanan Hu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Lu Yi
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Yeyi Yang
- Department of Medicine, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhixiang Wu
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Min Kong
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zhijuan Kang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China
| | - Zuocheng Yang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, People's Republic of China.
| |
Collapse
|
99
|
Hao XD, Liu JX, Zhang JS. Longevity factor FOXO3a: A potential therapeutic target for age-related ocular diseases. Life Sci 2024; 350:122769. [PMID: 38848943 DOI: 10.1016/j.lfs.2024.122769] [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: 02/28/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
The forkhead box protein O3 (FOXO3a) belongs to the subgroup O of the forkhead transcription factor family and plays an important role in regulating the aging process by participating in the regulation of various life processes, including cell cycle arrest, apoptosis, autophagy, oxidative stress, and DNA repair. The eye is an organ that is affected by aging earlier. However, the functional role and potential clinical applications of FOXO3a in age-related eye diseases have not received widespread attention and lacked comprehensive and clear clarification. In this review, we demonstrated the relationship between FOXO3a and visual system health, summarized the functional roles of FOXO3a in various eye diseases, and potential ocular-related therapies and drugs targeting FOXO3a in visual system diseases through a review and summary of relevant literature. This review indicates that FOXO3a is an important factor in maintaining the normal function of various tissues in the eye, and is closely related to the occurrence and development of ophthalmic-related diseases. Based on its vital role in the normal function of the visual system, FOXO3a has potential clinical application value in related ophthalmic diseases. At present, multiple molecules and drugs targeting FOXO3a have been reported to have the potential for the treatment of related ophthalmic diseases, but further clinical trials are needed. In conclusion, this review can facilitate us to grasp the role of FOXO3a in the visual system and provide new views and bases for the treatment strategy research of age-related eye diseases.
Collapse
Affiliation(s)
- Xiao-Dan Hao
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Jin-Xiu Liu
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Jing-Sai Zhang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
100
|
Lei Z, Pan C, Li F, Wei D, Ma Y. SGK1 promotes the lipid accumulation via regulating the transcriptional activity of FOXO1 in bovine. BMC Genomics 2024; 25:737. [PMID: 39080526 PMCID: PMC11290151 DOI: 10.1186/s12864-024-10644-0] [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: 03/11/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
OBJECTIVES Serum/glucocorticoid-inducible kinase 1 (SGK1) gene encodes a serine/threonine protein kinase that plays an essential role in cellular stress response and regulation of multiple metabolic processes. However, its role in bovine adipogenesis remains unknown. In this study, we aimed to clarify the role of SGK1 in bovine lipid accumulation and improvement of meat quality. METHODS Preadipocytes were induced to differentiation to detect the temporal expression pattern of SGK1. Heart, liver, lung, spleen, kidney, muscle and fat tissues were collected to detect its tissue expression profile. Recombinant adenovirus and the lentivirus were packaged for overexpression and knockdown. Oil Red O staining, quantitative real-time PCR, Western blot analysis, Yeast two-hybrid assay, luciferase assay and RNA-seq were performed to study the regulatory mechanism of SGK1. RESULTS SGK1 showed significantly higher expression in adipose and significantly induced expression in differentiated adipocytes. Furthermore, overexpression of SGK1 greatly promoted adipogenesis and inhibited proliferation, which could be shown by the remarkable increasement of lipid droplet, and the expression levels of adipogenic marker genes and cell cycle-related genes. Inversely, its knockdown inhibited adipogenesis and facilitated proliferation. Mechanistically, SGK1 regulates the phosphorylation and expression of two critical proteins of FoxO family, FOXO1/FOXO3. Importantly, SGK1 attenuates the transcriptional repression role of FOXO1 for PPARγ via phosphorylating the site S256, then promoting the bovine fat deposition. CONCLUSIONS SGK1 is a required epigenetic regulatory factor for bovine preadipocyte proliferation and differentiation, which contributes to a better understanding of fat deposition and meat quality improvement in cattle.
Collapse
Affiliation(s)
- Zhaoxiong Lei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Cuili Pan
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Fen Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Dawei Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| | - Yun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| |
Collapse
|