1
|
Yu YS, Kim IS, Baek SH. Decoding the dual role of autophagy in cancer through transcriptional and epigenetic regulation. FEBS Lett 2025. [PMID: 40346781 DOI: 10.1002/1873-3468.70060] [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: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/12/2025]
Abstract
Autophagy is a conserved catabolic process that is essential for maintaining cellular homeostasis by degrading and recycling damaged organelles and misfolded proteins. In cancer, autophagy exhibits a context-dependent dual role: In early stages, autophagy acts as a tumor suppressor by preserving genomic integrity and limiting oxidative stress. In advanced stages, autophagy supports tumor progression by facilitating metabolic adaptation, therapy resistance, immune evasion, and metastasis. This review highlights the molecular mechanisms underlying this dual function and focuses on the transcriptional and epigenetic regulation of autophagy in cancer cells. Key transcription factors, including the MiT/TFE family, FOXO family, and p53, as well as additional regulators, are discussed in the context of stress-responsive pathways mediated by mTORC1 and AMPK. A deeper understanding of the transcriptional and epigenetic regulation of autophagy in cancer is crucial for developing context-specific therapeutic strategies to either promote or inhibit autophagy depending on the cancer stage, thereby improving clinical outcomes in cancer treatment.
Collapse
Affiliation(s)
- Young Suk Yu
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Ik Soo Kim
- Department of Microbiology, Gachon University College of Medicine, Incheon, South Korea
| | - Sung Hee Baek
- Creative Research Initiatives Center for Epigenetic Code and Diseases, School of Biological Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
2
|
Yang F, Chen C, Chen R, Yang C, Liu Z, Wen L, Xiao H, Geng B, Xia Y. Unraveling the Potential of SGK1 in Osteoporosis: From Molecular Mechanisms to Therapeutic Targets. Biomolecules 2025; 15:686. [PMID: 40427579 PMCID: PMC12109298 DOI: 10.3390/biom15050686] [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: 03/15/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Osteoporosis (OP) is a prevalent metabolic bone disease, with several million cases of fractures resulting from osteoporosis worldwide each year. This phenomenon contributes to a substantial increase in direct medical expenditures and poses a considerable socioeconomic burden. Despite its prevalence, our understanding of the underlying mechanisms remains limited. Recent studies have demonstrated the involvement of serum glucocorticoid-regulated protein kinase 1 (SGK1) in multiple signaling pathways that regulate bone metabolism and its significant role in the development of osteoporosis. Therefore, it is of great significance to deeply explore the mechanism of SGK1 in osteoporosis and its therapeutic potential. In this paper, we present a comprehensive review of the structure and activation mechanism of SGK1, its biological function, the role of SGK1 in different types of osteoporosis, and the inhibitors of SGK1. The aim is to comprehensively assess the latest research progress with regards to SGK1's role in osteoporosis, clarify its role in the regulation of bone metabolism and its potential as a therapeutic target, and lay the foundation for the development of novel therapeutic strategies and personalized treatment in the future. Furthermore, by thoroughly examining the interactions between SGK1 and other molecules or signaling pathways, potential biomarkers may be identified, thereby enhancing the efficacy of early screening and intervention for osteoporosis.
Collapse
Affiliation(s)
- Fei Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics, Nanchong Central Hospital, Nanchong 637000, China
| | - Changshun Chen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Rongjin Chen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Chenghui Yang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Zirui Liu
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Lei Wen
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Hefang Xiao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (C.C.); (R.C.); (C.Y.); (Z.L.); (L.W.); (H.X.); (B.G.)
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
3
|
Wang Y, Wang B, Yan K, Liu L, Zhang J, Li Q, Huang Z, Wu KK, Liu S, Li J, Wei Q, Kwok HF, Luo S. Polysaccharides from Brasenia schreberi alleviated the toxicity induced by acrylamide on cells and Caenorhabditis elegans. Int J Biol Macromol 2025; 309:142831. [PMID: 40188917 DOI: 10.1016/j.ijbiomac.2025.142831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 04/02/2025] [Indexed: 05/10/2025]
Abstract
Acrylamide (AC), a widely used chemical compound in industrial production and the food industry, has been shown to exhibit reproductive toxicity, neurotoxicity, and genotoxicity. While no specific treatment has been developed to alleviate its harmful effects, identifying an effective therapeutic agent is necessary. In our previous work, polysaccharides from Brasenia schreberi (PSBS) demonstrated strong antioxidant activity, suggesting that PSBS may relieve AC-induced toxicity. In this study, we employed cellular models and Caenorhabditis elegans to evaluate the protective effects of PSBS against AC toxicity. The findings revealed that PSBS significantly reduced reactive oxygen species (ROS) levels and cell apoptosis through the activation of the MAPK signaling pathway. Furthermore, PSBS markedly improved the survival of C. elegans exposed to AC. Quantitative PCR (qPCR) analysis indicated that PSBS downregulated the insulin and PI3K/AKT pathways while upregulating the AMPK and MAPK pathways. Additionally, PSBS enhanced the expression of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), and glutathione S-transferase (GST) by activating skn-1 and daf-16. These results suggest that PSBS is a promising natural drug for mitigating acrylamide-induced toxicity.
Collapse
Affiliation(s)
- Yujie Wang
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Bangguo Wang
- The Third People's Hospital of Yibin, Yibin, China
| | - Kuan Yan
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Li Liu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Jian Zhang
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Qiang Li
- Food Safety Inspection Technology Center of Sichuan Market Supervision Administration, Chengdu, China
| | - Zhenglin Huang
- Food Safety Inspection Technology Center of Sichuan Market Supervision Administration, Chengdu, China
| | - Ka Kun Wu
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau
| | - Sicen Liu
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Junqiang Li
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Qin Wei
- Faculty of Agriculture, Forestry and Food Engineering, Yibin University, Yibin, China
| | - Hang Fai Kwok
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.
| | - Siyuan Luo
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau.
| |
Collapse
|
4
|
Asadi Y, Moundounga RK, Chakroborty A, Pokokiri A, Wang H. FOXOs and their roles in acute and chronic neurological disorders. Front Mol Biosci 2025; 12:1538472. [PMID: 40260403 PMCID: PMC12010098 DOI: 10.3389/fmolb.2025.1538472] [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: 12/02/2024] [Accepted: 02/10/2025] [Indexed: 04/23/2025] Open
Abstract
The forkhead family of transcription factors of class O (FOXOs) consisting of four functionally related proteins, FOXO1, FOXO3, FOXO4, and FOXO6, are mammalian homologs of daf-16 in Caenorhabditis elegans and were previously identified as tumor suppressors, oxidative stress sensors, and cell survival modulators. Under normal physiological conditions, FOXO protein activities are negatively regulated by phosphorylation via the phosphoinositide 3-kinase (PI3K)-Akt pathway, a well-known cell survival pathway: Akt phosphorylates FOXOs to inactivate their transcriptional activity by relocalizing FOXOs from the nucleus to the cytoplasm for degradation. However, under oxidative stress or absent the cellular survival drive of growth factors, FOXO proteins translocate to the nucleus and upregulate a series of target genes, thereby promoting cell growth arrest and cell death and altering mitochondrial homeostasis. FOXO gene expression is also regulated by other transcriptional factors such as p53 or autoregulation by their activities and end products. Here we summarize the structure, posttranslational modifications, and translocation of FOXOs linking to their transcriptional control of cellular functions, survival, and death, emphasizing their role in regulating the cellular response to some acute insults and chronic neurological disorders. This review will conclude with a brief section on potential therapeutic interventions that can be used to modulate FOXOs' activities when treating acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Yasin Asadi
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rozenn K. Moundounga
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Anand Chakroborty
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Augustina Pokokiri
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hongmin Wang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
5
|
Ma X, Lin Y, Zhang L, Huang Z, Zhang Y, Fu X, Li P. The dual missions of FoxO3a in inflammatory diseases: Regulation of antioxidant enzymes and involvement in programmed cell death. Int Immunopharmacol 2025; 151:114369. [PMID: 40031428 DOI: 10.1016/j.intimp.2025.114369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
The transcription factor FoxO3a plays a crucial role in the process of cells adapting to various stress conditions. Multiple post - translational modifications and epigenetic mechanisms work together to precisely regulate the activity of FoxO3a, influencing its subcellular localization, stability, interactions with other proteins, DNA - binding affinity, and transcriptional regulatory capacity. Under different chemical signal stimuli and subcellular environments, the activation of FoxO3a triggered by oxidative stress can initiate diverse transcriptional programs, which are essential for the body to resist oxidative damage. In the development and progression of inflammatory diseases, FoxO3a exerts an important function by regulating the expression levels of antioxidant enzymes and participating in key physiological processes such as programmed cell death. This article comprehensively reviews the structural characteristics, mechanism of action of FoxO3a, as well as its functions in regulating antioxidant enzymes and programmed cell death. The aim is to deeply explore the potential of FoxO3a as a potential therapeutic target for preventing and treating damages such as inflammatory diseases caused by cellular stress.
Collapse
Affiliation(s)
- Xiangli Ma
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yujie Lin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
6
|
Zhang Y, Wang M, Xu W, Zang H, Yan T, Wu T, Huang K, Chen D, Luo Q, Guo R, Qiu J. Analysis of the Expression Patterns of piRNAs in Response to Microsporidian Invasion in Midgut of Workers ( Apis cerana cerana). Int J Mol Sci 2025; 26:2402. [PMID: 40141043 PMCID: PMC11942432 DOI: 10.3390/ijms26062402] [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: 01/09/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Piwi-interacting RNAs (piRNAs) play an essential part in transposon suppression, DNA methylation, and antiviral responses. The current understanding of the roles of piRNAs in honeybees is very limited. This study aims to analyze the expression pattern and regulatory role of piRNAs in the Asian honeybee (Apis cerana) responding to infection by Nosema ceranae, based on previously gained small RNA-seq data. Here, 450 and 422 piRNAs were respectively identified in the midgut tissues of Apis cerana cerana workers at 7 and 10 days post-inoculation (dpi) with N. ceranae, including 539 non-redundant ones. Additionally, one up-regulated (piR-ace-1216942) and one down-regulated (piR-ace-776728) piRNA were detected in the workers' midgut at 7 dpi, targeting 381 mRNAs involved in 31 GO terms, such as metabolic processes, catalytic activity, and organelles, as well as 178 KEGG pathways, including lysosome, MAPK signaling pathway, and purine metabolism. A total of 35 up-regulated and 11 down-regulated piRNAs were screened from the workers' midgut at 10 dpi, targeting 13,511 mRNAs engaged in 50 GO terms, such as biological regulation, transporter activity, and membrane, as well as 389 KEGG pathways, including the JAK-STAT signaling pathway, Hippo signaling pathway, and nitrogen metabolism. Further analysis indicated that 28 differentially expressed piRNAs (DEpiRNAs) in the midgut at 10 dpi could target 299 mRNAs annotated to three cellular immune pathways (lysosome, endocytosis, and phagosome), while 24 DEpiRNAs could target 205 mRNAs relevant to four humoral immune pathways (FoxO, JAK-STAT, NF-κB, and MAPK signaling pathway). Through Sanger sequencing and RT-qPCR, the expression of six randomly selected DEpiRNAs was verified. Moreover, the dual-luciferase reporter gene assay confirmed the binding relationships between piR-ace-446232 and CRT as well as between piR-ace-1008436 and EGFR. Our findings not only contribute to enrich our understanding of the role of piRNAs in honeybees but also provide a basis for exploring the host response to N. ceranae infection mediated by piRNAs.
Collapse
Affiliation(s)
- Yiqiong Zhang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
| | - Mengyi Wang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
| | - Wenhua Xu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
| | - He Zang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
- Apitherapy Research Institute of Fujian Province, Fuzhou 350002, China
| | - Tizhen Yan
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
| | - Tao Wu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
| | - Kaifei Huang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
| | - Dafu Chen
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
- Apitherapy Research Institute of Fujian Province, Fuzhou 350002, China
| | - Qingming Luo
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
| | - Rui Guo
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
- Apitherapy Research Institute of Fujian Province, Fuzhou 350002, China
| | - Jianfeng Qiu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.Z.); (M.W.); (W.X.); (H.Z.); (T.W.); (K.H.); (D.C.)
- National & Local United Engineering Laboratory of Natural Biotoxin, Fuzhou 350002, China; (T.Y.); (Q.L.)
- Apitherapy Research Institute of Fujian Province, Fuzhou 350002, China
| |
Collapse
|
7
|
Wu K, Ge XX, Duan XF, Li JQ, Wang K, Chen QH, Huang ZM, Zhang WY, Wu Y, Li Q. Wip1 phosphatase activator QGC-8-52 specifically sensitizes p53-negative cancer cells to chemotherapy while protecting normal cells. Drug Resist Updat 2025; 79:101196. [PMID: 39787991 DOI: 10.1016/j.drup.2024.101196] [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: 04/15/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
PP2C serine-threonine phosphatase Wip1 plays an important role in normal tissue homeostasis, stress signaling and pathogenesis of various human diseases. It is an attractive drug target for cancer treatment and inhibition of its expression or activity constitute a novel therapeutic intervention strategy to prevent the development of various cancers. However, previous strategies for Wip1 suppression may be ineffective in cancers lacking p53. Here, we have characterized the activity of a novel Wip1 phosphatase activator, QGC-8-52, in preclinical models of breast malignancies. QGC-8-52 significantly sensitizes the cancer cell lines with p53 deletion to chemotherapeutic agents. This effect was mediated by the Wip1-FOXO3a interaction and subsequent dephosphorylation of Thr487 that resulted, in response to anticancer treatment, in enhancing the transcription activity of FOXO3a on the proapoptotic TRAIL gene. The sensitizing effect of Wip1 activation on chemotherapeutic drugs only targeted cancer cells lacking p53. The activation of Wip1 in normal cells provided protection from anticancer drug-induced apoptosis by reducing the strength of upstream signaling to p53. Therefore, during the treatment of anticancer drugs, the activated Wip1 phosphatase boosts the apoptosis of p53-negative tumors and protects normal tissues. Our findings may represent an effective and safe therapeutic strategy for cancers with p53 deletion.
Collapse
Affiliation(s)
- Ke Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charls Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA; School of Nursing, Wuhan University, Wuhan, 430071, China
| | - Xiao-Xiao Ge
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Xiao-Fan Duan
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China
| | - Jie-Qing Li
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qiao-Hong Chen
- Department of Chemistry, California State University, Fresno, 2555 E. San Ramon Avenue, M/S SB70, Fresno, CA 93740, USA
| | - Zhi-Min Huang
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | | | - Yong Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charls Drew University of Medicine and Science, David Geffen UCLA School of Medicine and UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
| | - Qun Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200123, China.
| |
Collapse
|
8
|
Luperchio AM, Salamango DJ. Defining the Protein Phosphatase 2A (PP2A) Subcomplexes That Regulate FoxO Transcription Factor Localization. Cells 2025; 14:342. [PMID: 40072071 PMCID: PMC11899004 DOI: 10.3390/cells14050342] [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: 01/30/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/15/2025] Open
Abstract
The family of forkhead box O (FoxO) transcription factors regulate cellular processes involved in glucose metabolism, stress resistance, DNA damage repair, and tumor suppression. FoxO transactivation activity is tightly regulated by a complex network of signaling pathways and post-translational modifications. While it has been well established that phosphorylation promotes FoxO cytoplasmic retention and inactivation, the mechanism underlying dephosphorylation and nuclear translocation is less clear. Here, we investigate the role of protein phosphatase 2A (PP2A) in regulating this process. We demonstrate that PP2A and AMP-activated protein kinase (AMPK) combine to regulate nuclear translocation of multiple FoxO family members following inhibition of metabolic signaling or induction of oxidative stress. Moreover, chemical inhibitor studies indicate that nuclear accumulation of FoxO proteins occurs through inhibition of nuclear export as opposed to promoting nuclear import as previously speculated. Functional, genetic, and biochemical studies combine to identify the PP2A complexes that regulate FoxO nuclear translocation, and the binding motif required. Mutating the FoxO-PP2A interface to enhance or diminish PP2A binding alters nuclear translocation kinetics accordingly. Together, these studies shed light on the molecular mechanisms regulating FoxO nuclear translocation and provide insights into how FoxO regulation is integrated with metabolic and stress-related stimuli.
Collapse
Affiliation(s)
| | - Daniel J. Salamango
- Department of Microbiology, Immunology, and Molecular Genetics, UT Health Science Center, San Antonio, TX 78229, USA;
| |
Collapse
|
9
|
Oyama T, Brashears CB, Rathore R, Benect-Hamilton H, Caldwell KE, Dirckx N, Hawkins WG, Van Tine BA. PHGDH inhibition and FOXO3 modulation drives PUMA-dependent apoptosis in osteosarcoma. Cell Death Dis 2025; 16:89. [PMID: 39934141 PMCID: PMC11814296 DOI: 10.1038/s41419-025-07378-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/23/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025]
Abstract
Osteosarcoma is a bone cancer that has been found to be metabolically dependent on the conversion of glucose to serine through the rate-limiting enzyme 3-phosphoglycerate dehydrogenase (PHGDH). The upregulation of PHGDH has been correlated with poor patient survival, and the inhibition of the serine synthesis pathway using targeted small-molecule inhibition of PHGDH induces a rapid metabolic adaptation that prevents cell death due to pro-survival signaling through the mammalian target of rapamycin complex 1 (mTORC1) pathway. Here, PHGDH inhibition in combination with mTORC1 signaling modulation for the treatment of osteosarcoma was evaluated. When combined with PHGDH inhibition, several non-rapalog inhibitors of mTORC1 activated Forkhead box O (FOXO) transcription factor 3 (FOXO3), a transcription factor associated with various cellular processes driving apoptosis. The activation of FOXO3 led to transcriptional activation of the pro-apoptotic gene p53 upregulated modulator of apoptosis (PUMA), inducing apoptosis when combined with PHGDH inhibition. These data suggest a path for the clinical development of PHGDH inhibitors in conjunction with mTORC1 pathway modulators in osteosarcoma.
Collapse
Affiliation(s)
- Toshinao Oyama
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, St. Louis, MO, USA.
| | - Caitlyn B Brashears
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Richa Rathore
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Heather Benect-Hamilton
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | - Katharine E Caldwell
- Department of Surgery, Division of Hepatobiliary Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | - Naomi Dirckx
- Department of Orthopedics, Washington University in St. Louis, St. Louis, MO, USA
| | - William G Hawkins
- Department of Surgery, Division of Hepatobiliary Surgery, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, St. Louis, MO, USA
| | - Brian A Van Tine
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis, St. Louis, MO, USA.
- Siteman Cancer Center, St. Louis, MO, USA.
- Department of Pediatric Hematology/Oncology, St Louis Children's Hospital, St Louis, MO, USA.
| |
Collapse
|
10
|
Pei S, Zhang D, Li Z, Liu J, Li Z, Chen J, Xie Z. The Role of the Fox Gene in Breast Cancer Progression. Int J Mol Sci 2025; 26:1415. [PMID: 40003882 PMCID: PMC11855465 DOI: 10.3390/ijms26041415] [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: 11/26/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Forkhead box (FOX) genes are a family of transcription factors that participate in many biological activities, from early embryogenesis to the formation of organs, and from regulation of glucose metabolism to regulation of longevity. Given the extensive influence in the multicellular process, FOX family proteins are responsible for the progression of many types of cancers, especially lung cancer, breast cancer, prostate cancer, and other cancers. Breast cancer is the most common cancer among women, and 2.3 million women were diagnosed in 2020. So, various drugs targeting the FOX signaling pathway have been developed to inhibit breast cancer progression. While the role of the FOX family gene in cancer development has not received enough attention, discovering more potential drugs targeting the FOX signaling pathway is urgently demanded. Here, we review the main members in the FOX gene family and summarize their signaling pathway, including the regulation of the FOX genes and their effects on breast cancer progression. We hope this review will emphasize the understanding of the role of the FOX gene in breast cancer and inspire the discovery of effective anti-breast cancer medicines targeting the FOX gene in the future.
Collapse
Affiliation(s)
- Shaoxuan Pei
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Dechun Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhuohan Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jinkai Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Ziyi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Jianrui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China
| | - Zhenzhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (S.P.); (D.Z.); (Z.L.); (J.L.); (Z.L.); (J.C.)
| |
Collapse
|
11
|
Chang M, Liu R, Chen B, Xu J, Wang W, Ji Y, Gao Z, Liu B, Yao X, Sun H, Xu F, Shen Y. hBMSC-EVs alleviate weightlessness-induced skeletal muscle atrophy by suppressing oxidative stress and inflammation. Stem Cell Res Ther 2025; 16:46. [PMID: 39901193 PMCID: PMC11792267 DOI: 10.1186/s13287-025-04175-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Muscle disuse and offloading in microgravity are likely the primary factors mediating spaceflight-induced muscle atrophy, for which there is currently no effective treatment other than exercise. Extracellular vesicles derived from bone marrow mesenchymal stem cells (BMSC-EVs) possess anti-inflammatory and antioxidant properties, offering a potential strategy for combating weightless muscular atrophy. METHODS In this study, human BMSCs-EVs (hBMSC-EVs) were isolated using super-centrifugation and characterized. C2C12 myotube nutrition-deprivation and mice tail suspension models were established. Subsequently, the diameter of C2C12 myotubes, Soleus mass, cross-sectional area (CSA) of muscle fibers, and grip strength in mice were assessed to investigate the impact of hBMSC-EVs on muscle atrophy. Immunostaining, transmission electron microscopy observation, and western blot analysis were employed to assess the impact of hBMSC-EVs on muscle fiber types, ROS levels, inflammation, ubiquitin-proteasome system activity, and autophagy lysosome pathway activation in skeletal muscle atrophy. RESULTS The active hBMSC-EVs can be internalized by C2C12 myotubes and skeletal muscle. hBMSC-EVs can effectively reduce C2C12 myotube atrophy caused by nutritional deprivation, with a concentration of 10 × 108 particles/mL showing the best effect (P < 0.001). Additionally, hBMSC-EVs can down-regulate the protein levels associated with UPS and oxidative stress. Moreover, intravenous administration of hBMSC-EVs at a concentration of 1 × 1010 particles/mL can effectively reverse the reduction in soleus mass (P < 0.001), CSA (P < 0.01), and grip strength (P < 0.001) in mice caused by weightlessness. They demonstrate the ability to inhibit protein degradation mediated by UPS and autophagy lysosome pathway, along with the suppression of oxidative stress and inflammatory responses. Furthermore, hBMSC-EVs impede the transition of slow muscle fibers to fast muscle fibers via upregulation of Sirt1 and PGC-1α protein levels. CONCLUSIONS Our findings indicate that hBMSC-EVs are capable of inhibiting excessive activation of the UPS and autophagy lysosome pathway, suppressing oxidative stress and inflammatory response, reversing muscle fiber type transformation, effectively delaying hindlimb unloading-induced muscle atrophy and enhancing muscle function. Our study has further advanced the understanding of the molecular mechanism underlying muscle atrophy in weightlessness and has demonstrated the protective effect of hBMSC-EVs on muscle atrophy.
Collapse
Affiliation(s)
- Mengyuan Chang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Ruiqi Liu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Bingqian Chen
- Department of Orthopedics, First People's Hospital of Changshu City, Changshu Hospital Affiliated to Soochow University, Changshu, 215500, Jiangsu Province, People's Republic of China
| | - Jin Xu
- Department of Basic Medicine, Kangda College of Nanjing Medical University, Lianyungang, 222000, Jiangsu Province, People's Republic of China
| | - Wei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Yanan Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China.
- Research and Development Center for E-Learning, Ministry of Education, Beijing, People's Republic of China.
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu Province, People's Republic of China.
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
12
|
Monaghan-Benson E, Aureille J, Guilluy C. ECM stiffness regulates lung fibroblast survival through RasGRF1-dependent signaling. J Biol Chem 2025; 301:108161. [PMID: 39793891 PMCID: PMC11835592 DOI: 10.1016/j.jbc.2025.108161] [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: 07/03/2024] [Revised: 12/20/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Extracellular matrix stiffness is one of the multiple mechanical signals that alter cellular behavior. During studies exploring the effect of matrix rigidity on lung fibroblast survival, we discovered that enhanced survival on stiff substrates is dependent on elevated Ras activity, owing to the activation of the guanine nucleotide exchange factor, RasGRF1. Mechanistically, we found that the increased Ras activity lead to the activation of both the AKT and ERK pathways. Pharmacological inhibition of AKT or ERK signaling attenuates the elevated survival observed on stiff substrates. AKT signaling regulates the phosphorylation and inactivation of the transcription factor FOXO3a. RNAi experiments demonstrate that FOXO3a activity is critical for the cell death observed on soft substrates. Additionally, downregulation of FOXO3a activity on stiff substrate leads to the degradation of the proapoptotic protein Bim. Depletion of Bim increased the survival of cells on soft substrates. Together, our data show that enhanced matrix stiffness activates a RasGRF1/Ras signaling cascade that regulates the activity of AKT and ERK-dependent FOXO3a and Bim expression to alter cell survival.
Collapse
Affiliation(s)
- Elizabeth Monaghan-Benson
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Julien Aureille
- Institute for Advanced Biosciences Centre de recherche UGA, INSERM U1209, CNRS UMR, Grenoble, France
| | - Christophe Guilluy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
13
|
de Korte D, Hoekstra M. Protein Arginine Methyltransferase 1: A Multi-Purpose Player in the Development of Cancer and Metabolic Disease. Biomolecules 2025; 15:185. [PMID: 40001488 PMCID: PMC11852820 DOI: 10.3390/biom15020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Protein arginine methyltransferase 1 (PRMT1) is the main PRMT family member involved in the formation of monomethylarginine and asymmetric dimethylarginine on its protein substrates. Many protein substrates of PRMT1 are key mediators of cell proliferation and oncogenesis. As such, the function of PRMT1 has been most prominently investigated in the context of cancer development. However, recent in vitro and in vivo studies have highlighted that PRMT1 may also promote metabolic disorders. With the current review, we aim to present an in-depth overview of how PRMT1 influences epigenetic modulation, transcriptional regulation, DNA damage repair, and signal transduction in cancer. Furthermore, we summarize the current knowledge regarding the role of PRMT1 in metabolic reprogramming, lipid metabolism, and glucose metabolism and describe the association of PRMT1 with numerous metabolic pathologies such as obesity, liver disease, and type 2 diabetes. It has become apparent that inhibiting the function of PRMT1 will likely serve as the most beneficial therapeutic approach, since several PRMT1 inhibitors have already been shown to exert positive effects on both cancer and metabolic disease in preclinical settings. However, pharmacological PRMT1 inhibition has not yet been shown to be therapeutically effective in clinical studies.
Collapse
Affiliation(s)
| | - Menno Hoekstra
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Gorlaeus Laboratories, Einsteinweg 55, 2333 CC Leiden, The Netherlands;
| |
Collapse
|
14
|
Huang Z, Jing H, Pan Y, Cai H, Zhang W, Zhu J, Zhang N, Wu D, Xu W, Qiu H, Bao H, Li G, Ning J, Xian B, Gao S. L-Theanine Extends the Lifespan of Caenorhabditis elegans by Reducing the End Products of Advanced Glycosylation. Foods 2025; 14:221. [PMID: 39856887 PMCID: PMC11764849 DOI: 10.3390/foods14020221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
L-theanine, a non-protein amino acid naturally occurring in tea leaves, is recognized for its antioxidant, anti-inflammatory, and neuroprotective properties. Despite its known benefits, the mechanisms by which L-theanine influences lifespan extension remain poorly understood. This study investigated the effects of L-theanine on the lifespan of Caenorhabditis elegans and explored the underlying mechanisms. Our findings indicate that L-theanine significantly diminishes the accumulation of advanced glycation end products (AGEs), which are biomarkers closely linked to aging and age-related diseases. Through an AGE-level analysis, we observed that L-theanine, when administered during early adulthood, notably extended the lifespan of Caenorhabditis elegans under both normal and high-glucose-induced stress conditions. L-theanine enhanced the lifespan under typical conditions and provided protective effects against high-glucose-induced stress. A further analysis demonstrated that L-theanine extends the lifespan of Caenorhabditis elegans by modulating the DAF-2/DAF-16 insulin-like signaling pathway and reducing the accumulation of advanced glycation end products (AGEs). In summary, this study identified L-theanine as a potential anti-aging intervention that extends the lifespan by reducing AGE accumulation and regulating insulin-like signaling pathways. These findings provide new insights for developing anti-aging strategies and lay the groundwork for further research on the potential benefits of L-theanine in mammals. Future studies could explore the molecular mechanisms, test L-theanine in mammalian models, and assess the long-term side effects.
Collapse
Affiliation(s)
- Zhihang Huang
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Haiming Jing
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
| | - Yan Pan
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Hongxia Cai
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Wenjing Zhang
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
| | - Jingyuan Zhu
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Nan Zhang
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
| | - Dan Wu
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Wentao Xu
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Hexiang Qiu
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Huihui Bao
- NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Science Research Unit, China National Center for Food Safety Risk Assessment, Beijing 100022, China;
| | - Guojun Li
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Junyu Ning
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
| | - Bo Xian
- Laboratory of Aging Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610056, China; (Y.P.); (H.C.); (J.Z.); (D.W.); (W.X.); (H.Q.)
| | - Shan Gao
- Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China; (Z.H.); (H.J.); (W.Z.); (N.Z.); (G.L.); (J.N.)
| |
Collapse
|
15
|
Kaur P, Khan H, Grewal AK, Dua K, Singh SK, Gupta G, Singh TG. Exploring Therapeutic Strategies: The Relationship between Metabolic Disorders and FOXO Signalling in Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:196-207. [PMID: 39473249 DOI: 10.2174/0118715273321002240919102841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/28/2024] [Accepted: 08/08/2024] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease is an ailment that is linked with the degeneration of the brain cells, and this illness is the main cause of dementia. Metabolic stress affects the activity of the brain in AD via FOXO signaling. The occurrence of AD will significantly surge as the world's population ages, along with lifestyle changes perceived in current decades, indicating a main contributor to such augmented prevalence. Similarly, metabolic disorders of current adulthood, such as obesity, stroke, and diabetes mellitus, have been observed as the risk-causing factors of AD. Environmental influences induce genetic mutations that result in the development of several diseases. Metabolic disorders develop when individuals are exposed to an environment where food is easily accessible and requires minimal energy expenditure. Obesity and diabetes are among the most significant worldwide health concerns. Obesity arises because of an imbalance between the amount of energy consumed and the amount of energy expended, which is caused by both behavioral and physiological factors. Obesity, insulin resistance syndrome, hypertension, and inflammation are factors that contribute to the worldwide risk of developing diabetes mellitus and neurodegenerative diseases. FOXO transcription factors are preserved molecules that play an important part in assorted biological progressions, precisely in aging as well as metabolism. Apoptosis, cell division and differentiation, oxidative stress, metabolism, and lifespan are among the physiological processes that the FOXO proteins are adept at controlling. In this review, we explored the correlation between signaling pathways and the cellular functions of FOXO proteins. We have also summarized the intricate role of FOXO in AD, with a focus on metabolic stress, and discussed the prospect of FOXO as a molecular link between AD and metabolic disorders.
Collapse
Affiliation(s)
- Parneet Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Heena Khan
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Amarjot Kaur Grewal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sachin Kumar Singh
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, 302017, Mahal Road, Jaipur, India
- Centre for Transdisciplinary Research, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
- Department of Pharmacology, School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
| | - Thakur Gurjeet Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| |
Collapse
|
16
|
Shi JH, Shi YB, Qiu ST, Song Y. Role of the Transcription Factor FoxO in Type 2 Diabetes and Its Complications. Crit Rev Eukaryot Gene Expr 2025; 35:85-103. [PMID: 39964972 DOI: 10.1615/critreveukaryotgeneexpr.2025057309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
FoxO proteins represent a subfamily of the forkhead box family (Fox) superfamily of proteins. It is involved in cell proliferation, differentiation, oxidative stress, apoptosis as well as tumors and metabolic disorders by regulating cellular functions. This paper aims to summarize the role of the transcription factor FoxO in type 2 diabetes and its complications, which may add to the potential of FoxO as a therapeutic target for future research. The transcription factor FoxO is expressed in various tissues and participates in various bodily functions including cell proliferation, differentiation, apoptosis, tumor therapy, and metabolic processes, playing a crucial role in the human body. FoxO plays a positive role in attenuating oxidative stress, inflammation, and metabolic disorders, which are the main causes of type 2 diabetes and its complications. FoxO plays an important role in the regulation of type 2 diabetes and its complications, and more precise targeting studies of FoxO will help to prevent, regulate, and treat diabetes-related diseases.
Collapse
Affiliation(s)
- Jing Hui Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yi Biao Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Si Tian Qiu
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, 18# Chaowang Road, Hangzhou City, Zhejiang Province 310014, P.R. China
| |
Collapse
|
17
|
Kumar V, Lakshman PKC, Kamariah N. Fluorescence Polarization Method to Assess the FOXO Phosphopeptide Interaction with 14-3-3. Methods Mol Biol 2025; 2871:99-106. [PMID: 39565581 DOI: 10.1007/978-1-0716-4217-7_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] [Indexed: 11/21/2024]
Abstract
Fluorescence polarization (FP) is a reliable and straightforward mix-and-read assay to quantify the interaction of a protein-peptide pair. Labeling the peptide with a suitable fluorophore enables the distinction between the protein-bound and protein-unbound states of a peptide by a change in the emitted light's polarization. Hereby, we present the method to establish the binding affinity for FOXO3a phosphopeptide with 14-3-3ɛ protein using FP. With the advantages of lower sample consumption and faster read-outs, FP validates the interaction between the pairs and can also be used to develop a competitive assay to identify and validate the novel inhibitors for the interaction pair.
Collapse
Affiliation(s)
- Vikrant Kumar
- Centre for Chemical Biology and Therapeutics, Institute for Stem Cell Science and Regenerative Medicine and National Centre for Biological Sciences-TIFR, Bangalore, India
| | - Puneeth Kumar Chunchagatta Lakshman
- Centre for Chemical Biology and Therapeutics, Institute for Stem Cell Science and Regenerative Medicine and National Centre for Biological Sciences-TIFR, Bangalore, India
| | - Neelagandan Kamariah
- Centre for Chemical Biology and Therapeutics, Institute for Stem Cell Science and Regenerative Medicine and National Centre for Biological Sciences-TIFR, Bangalore, India.
| |
Collapse
|
18
|
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: 0] [Impact Index Per Article: 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
|
19
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
20
|
Xu J, Yuan Z, Zhao H, Wu X, Cai N, Ma T, Tang B, Chen G, Wang S. RNAi-Mediated FoxO Silencing Inhibits Reproduction in Locusta migratoria. INSECTS 2024; 15:891. [PMID: 39590490 PMCID: PMC11594837 DOI: 10.3390/insects15110891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/20/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024]
Abstract
FoxO is a downstream target gene of cellular nutrient and growth factors, oxidative stress responses, and insulin signaling pathways. It play a crucial role in insect growth, development, and reproduction. Locusta migratoria is a significant agricultural pest; therefore, the identification of novel control targets for its management is of significant importance. After injecting dsRNA to interfere with FoxO expression, we observed changes in the reproduction-related gene expression and ovary development through RT-qPCR and morphological observation. Simultaneously, the trehalose and glycogen contents were measured following RNAi. The results demonstrate that interference with FoxO significantly downregulates key genes in the Hippo pathway and Notch gene expression. In terms of carbohydrate metabolism, the trehalose content decreases significantly while the glycogen content increases markedly after FoxO silencing. Additionally, FoxO silencing considerably inhibits reproductive-related gene expression, resulting in delayed ovarian development. These findings indicate that FoxO regulates L. migratoria reproduction through the Hippo signaling pathway: when impaired, the reproductive capacity function declines. In addition, FoxO-mediated energy mobilization is involved in the regulation of egg production. These results indicate that the RNAi of FoxO may be a useful control strategy against L. migratoria.
Collapse
Affiliation(s)
- Jiaying Xu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Zeming Yuan
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Huazhang Zhao
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Xinru Wu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Nina Cai
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Tingting Ma
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Bin Tang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| | - Gongxing Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Shigui Wang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; (J.X.); (Z.Y.); (H.Z.); (X.W.); (N.C.); (T.M.); (B.T.)
| |
Collapse
|
21
|
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
|
22
|
Zhou R, Barnes K, Gibson S, Fillmore N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1162-H1173. [PMID: 39269450 DOI: 10.1152/ajpheart.00001.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Regulation of energy metabolism is pivotal in the development of cardiovascular diseases. Dysregulation in mitochondrial fatty acid oxidation has been linked to cardiac lipid accumulation and diabetic cardiomyopathy. Sirtuin 1 (SIRT1) is a deacetylase that regulates the acetylation of various proteins involved in mitochondrial energy metabolism. SIRT1 mediates energy metabolism by directly and indirectly affecting multiple aspects of mitochondrial processes, such as mitochondrial biogenesis. SIRT1 interacts with essential mitochondrial energy regulators such as peroxisome proliferator-activated receptor-α (PPARα), PPARγ coactivator-1α, estrogen-related receptor-α, and their downstream targets. Apart from that, SIRT1 regulates additional proteins, including forkhead box protein O1 and AMP-activated protein kinase in cardiac disease. Interestingly, studies have also shown that the expression of SIRT1 plays a dual-edged role in energy metabolism. Depending on the physiological state, SIRT1 expression can be detrimental or protective. This review focuses on the molecular pathways through which SIRT1 regulates energy metabolism in cardiovascular diseases. We will review SIRT1 and discuss its role in cardiac energy metabolism and its benefits and detrimental effects in heart disease.
Collapse
Affiliation(s)
- Redemptor Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Kaleb Barnes
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Savannah Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
23
|
Schade AE, Perurena N, Yang Y, Rodriguez CL, Krishnan A, Gardner A, Loi P, Xu Y, Nguyen VTM, Mastellone GM, Pilla NF, Watanabe M, Ota K, Davis RA, Mattioli K, Xiang D, Zoeller JJ, Lin JR, Morganti S, Garrido-Castro AC, Tolaney SM, Li Z, Barbie DA, Sorger PK, Helin K, Santagata S, Knott SRV, Cichowski K. AKT and EZH2 inhibitors kill TNBCs by hijacking mechanisms of involution. Nature 2024; 635:755-763. [PMID: 39385030 PMCID: PMC11578877 DOI: 10.1038/s41586-024-08031-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/06/2024] [Indexed: 10/11/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and has the highest rate of recurrence1. The predominant standard of care for advanced TNBC is systemic chemotherapy with or without immunotherapy; however, responses are typically short lived1,2. Thus, there is an urgent need to develop more effective treatments. Components of the PI3K pathway represent plausible therapeutic targets; more than 70% of TNBCs have alterations in PIK3CA, AKT1 or PTEN3-6. However, in contrast to hormone-receptor-positive tumours, it is still unclear whether or how triple-negative disease will respond to PI3K pathway inhibitors7. Here we describe a promising AKT-inhibitor-based therapeutic combination for TNBC. Specifically, we show that AKT inhibitors synergize with agents that suppress the histone methyltransferase EZH2 and promote robust tumour regression in multiple TNBC models in vivo. AKT and EZH2 inhibitors exert these effects by first cooperatively driving basal-like TNBC cells into a more differentiated, luminal-like state, which cannot be effectively induced by either agent alone. Once TNBCs are differentiated, these agents kill them by hijacking signals that normally drive mammary gland involution. Using a machine learning approach, we developed a classifier that can be used to predict sensitivity. Together, these findings identify a promising therapeutic strategy for this highly aggressive tumour type and illustrate how deregulated epigenetic enzymes can insulate tumours from oncogenic vulnerabilities. These studies also reveal how developmental tissue-specific cell death pathways may be co-opted for therapeutic benefit.
Collapse
Affiliation(s)
- Amy E Schade
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Naiara Perurena
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yoona Yang
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Carrie L Rodriguez
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anjana Krishnan
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Alycia Gardner
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Patrick Loi
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yilin Xu
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Van T M Nguyen
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - G M Mastellone
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Natalie F Pilla
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marina Watanabe
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Keiichi Ota
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rachel A Davis
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kaia Mattioli
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dongxi Xiang
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason J Zoeller
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefania Morganti
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ana C Garrido-Castro
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara M Tolaney
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Zhe Li
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter K Sorger
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Kristian Helin
- Division of Cancer Biology, Institute of Cancer Research, London, UK
| | - Sandro Santagata
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Simon R V Knott
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Karen Cichowski
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Tumolo MR, Bodini A, Bagordo F, Leo CG, Mincarone P, De Matteis E, Sabina S, Grassi T, Scoditti E. MiRNA Expression in Long-Distance Runners with Musculoskeletal Pain: Implications for Pain Pathophysiology. Biomedicines 2024; 12:2494. [PMID: 39595060 PMCID: PMC11591860 DOI: 10.3390/biomedicines12112494] [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: 09/10/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Background: miRNAs are short, non-coding RNAs whose deregulation has been shown in painful processes, including musculoskeletal pain. This condition, which causes disability, impacts quality of life, and contributes to substantial healthcare costs, is also a critical issue in sports. In this case-control study, we evaluated the expression of four miRNAs involved in inflammation in runners with musculoskeletal pain and elucidated their functions and pathophysiological implications. Methods: A total of 17 runners with musculoskeletal pain and 17 age- and sex-matched runners without pain participated in this study. The levels of the miRNAs were evaluated by qRT-PCR. Bioinformatic tools were employed to identify the target genes and biological processes regulated by these miRNAs. Results: Compared to the controls, the runners with musculoskeletal pain exhibited significantly higher plasma levels of miR-133b (p = 0.02), miR-155-5p (p = 0.003) and let-7a-5p (p = 0.02). Multivariable regression analysis indicated that these three miRNAs exhibit a positive correlation (p < 0.05) with the presence of musculoskeletal pain, adjusted for age. Bioinformatic analysis suggested that the miRNAs hub genes are involved in regulatory processes, neuroinflammatory pathways, and human diseases that are associated with pain pathology. Conclusions: These results enhance our understanding of the potential role of miR-133b, miR-155-5p and let-7a-5p in pain-associated biological processes. The miRNA-mediated negative regulation of genes identified could explain the inflammatory and tissue repair processes in this population. Further studies are needed to confirm and validate the role of these miRNAs in painful conditions, especially considering the significant public health implications of managing inflammatory pain in sports.
Collapse
Affiliation(s)
- Maria Rosaria Tumolo
- Department of Biological & Environmental Sciences & Technology, University of Salento, 73100 Lecce, Italy;
- Branch of Lecce, National Research Council, Institute of Clinical Physiology, 73100 Lecce, Italy or (C.G.L.); (E.S.)
| | - Antonella Bodini
- National Research Council, Institute for Applied Mathematics & Information Technologies ‘E. Magenes’, 20133 Milan, Italy;
| | - Francesco Bagordo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy;
| | - Carlo Giacomo Leo
- Branch of Lecce, National Research Council, Institute of Clinical Physiology, 73100 Lecce, Italy or (C.G.L.); (E.S.)
- MOVE-mentis s.r.l., 47522 Cesena, Italy;
| | - Pierpaolo Mincarone
- MOVE-mentis s.r.l., 47522 Cesena, Italy;
- Research Unit of Brindisi, National Research Council, Institute for Research on Population & Social Policies, 72100 Brindisi, Italy
| | | | - Saverio Sabina
- Branch of Lecce, National Research Council, Institute of Clinical Physiology, 73100 Lecce, Italy or (C.G.L.); (E.S.)
- MOVE-mentis s.r.l., 47522 Cesena, Italy;
| | - Tiziana Grassi
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy;
| | - Egeria Scoditti
- Branch of Lecce, National Research Council, Institute of Clinical Physiology, 73100 Lecce, Italy or (C.G.L.); (E.S.)
| |
Collapse
|
25
|
Pucci G, Savoca G, Iacoviello G, Russo G, Forte GI, Cavalieri V. Curcumin's Radioprotective Effects on Zebrafish Embryos. Antioxidants (Basel) 2024; 13:1281. [PMID: 39594423 PMCID: PMC11590968 DOI: 10.3390/antiox13111281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Radiation modifiers are largely studied for their contribution to enlarging the treatment window. Curcumin is already known for its antioxidant properties; however, its role as a radioprotector in preclinical studies is affected by the well-known low absorption and bioavailability of curcumin. In this study, curcumin's radioprotection ability has been evaluated in zebrafish larvae, by taking advantage of quantifying curcumin absorption and evaluating its fluorescence in transparent embryos. A curcumin range of 1-10 μM was tested to select the non-toxic concentrations to be used for a pre-treatment of photon beam irradiation using a 2-15 Gy range of doses. The post-treatment analysis within 120 h post-fertilization (hpf) included an assessment of mortality and malformation rates and behavioral and gene expression analysis. A total of 2.5 and 5 μM of curcumin pre-treatment showed a radioprotective role, significantly reducing the frequency of embryo malformations and damaged entities. This sparing effect disappeared using 15 Gy, showing the radiation effect's prevalence. Gene expression analysis reconducted this radioprotective ability for antioxidant gene network activation. The curcumin-induced activation of the antioxidant gene network promoted radioprotection in zebrafish.
Collapse
Affiliation(s)
- Gaia Pucci
- Institute of Bioimaging and Complex Biological Systems (IBSBC)—National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.R.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld. 16, 90128 Palermo, Italy
| | - Gaetano Savoca
- Radiation Oncology Unit, ARNAS-Civico Hospital, 90100 Palermo, Italy; (G.S.); (G.I.)
| | - Giuseppina Iacoviello
- Radiation Oncology Unit, ARNAS-Civico Hospital, 90100 Palermo, Italy; (G.S.); (G.I.)
| | - Giorgio Russo
- Institute of Bioimaging and Complex Biological Systems (IBSBC)—National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.R.)
| | - Giusi I. Forte
- Institute of Bioimaging and Complex Biological Systems (IBSBC)—National Research Council (CNR), Cefalù Secondary Site, C/da Pietrapollastra-Pisciotto, 90015 Cefalù, Italy; (G.P.); (G.R.)
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld. 16, 90128 Palermo, Italy
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STeBiCeF), University of Palermo, Viale delle Scienze Bld. 16, 90128 Palermo, Italy
- Zebrafish Laboratory, Advanced Technologies Network (ATeN) Center, University of Palermo, Viale delle Scienze Bld. 16, 90128 Palermo, Italy
| |
Collapse
|
26
|
Berk Ş. Insulin and IGF-1 extend the lifespan of Caenorhabditis elegans by inhibiting insulin/insulin-like signaling and mTOR signaling pathways: C. elegans - Focused cancer research. Biochem Biophys Res Commun 2024; 729:150347. [PMID: 38976945 DOI: 10.1016/j.bbrc.2024.150347] [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: 04/28/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
The mutations in Caenorhabditis elegans (C. elegans) that extend lifespan slow down aging by interfering with several signaling pathways, including the insulin/IGF-1 signaling (IIS) pathway, AMP-activated protein kinase (AMPK), and mechanistic target of rapamycin (mTOR). The tumor suppressor pRb (retinoblastoma protein) is believed to be involved in almost all human cancers. Lin-35, the C. elegans orthologue of the tumor suppressor pRb, was included in the study to explore the effects of insulin and IGF-1 because it has been linked to cancer-related pRb function in mammals and exhibits a tumor suppressor effect by inhibiting mTOR or IIS signaling. According to our results, IGF-1 or insulin increased the lifespan of lin-35 worms compared to N2 worms by increasing fertilization efficiency, also causing a significant increase in body size. It was concluded that the expression of daf-2 and rsks-1 decreased after insulin or IGF-1 administration, thus extending the lifespan of C. elegans lin-35 worms through both IIS and mTOR-dependent mechanisms. This suggests that it was mediated by the combined effect of the TOR and IIS pathways. These results, especially obtained in cancer-associated mutant lin-35 worms, will be useful in elucidating the C. elegans cancer model in the future.
Collapse
Affiliation(s)
- Şeyda Berk
- Department of Molecular Biology and Genetics, Faculty of Science, Sivas Cumhuriyet University, Sivas, 58140, Turkey; Advanced Technology Research and Application Center (CUTAM), Sivas Cumhuriyet University, Sivas, 58140, Turkey.
| |
Collapse
|
27
|
Bhandari S, Kyrrestad I, Simón-Santamaría J, Li R, Szafranska KJ, Dumitriu G, Sánchez Romano J, Smedsrød B, Sørensen KK. Mouse liver sinusoidal endothelial cell responses to the glucocorticoid receptor agonist dexamethasone. Front Pharmacol 2024; 15:1377136. [PMID: 39439887 PMCID: PMC11494038 DOI: 10.3389/fphar.2024.1377136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) which make up the fenestrated wall of the hepatic sinusoids, are active scavenger cells involved in blood waste clearance and liver immune functions. Dexamethasone is a synthetic glucocorticoid commonly used in the clinic and as cell culture supplement. However, the response is dependent on tissue, cell type, and cell state. The aim of this study was to investigate the effect of dexamethasone on primary mouse LSECs (C57BL/6J); their viability (live-dead, LDH release, caspase 3/7 assays), morphology (scanning electron microscopy), release of inflammatory markers (ELISA), and scavenging functions (endocytosis assays), and associated biological processes and pathways. We have characterized and catalogued the proteome of LSECs cultured for 1, 10, or 48 h to elucidate time-dependent and dexamethasone-specific cell responses. More than 6,000 protein IDs were quantified using tandem mass tag technology and advanced mass spectrometry (synchronous precursor selection multi-notch MS3). Enrichment analysis showed a culture-induced upregulation of stress and inflammatory markers, and a significant shift in cell metabolism already at 10 h, with enhancement of glycolysis and concomitant repression of oxidative phosphorylation. At 48 h, changes in metabolic pathways were more pronounced with dexamethasone compared to time-matched controls. Dexamethasone repressed the activation of inflammatory pathways (IFN-gamma response, TNF-alpha signaling via NF-kB, Cell adhesion molecules), and culture-induced release of interleukin-6, VCAM-1, and ICAM-1, and improved cell viability partly through inhibition of apoptosis. The mouse LSECs did not proliferate in culture. Dexamethasone treated cells showed upregulation of xanthine dehydrogenase/oxidase (Xdh), and the transcription regulator Foxo1. The drug further delayed but did not block the culture-induced loss of LSEC fenestration. The LSEC capacity for endocytosis was significantly reduced at 48 h, independent of dexamethasone, which correlated with diminished expression of several scavenger receptors and C-type lectins and altered expression of proteins in the endocytic machinery. The glucocorticoid receptor (NR3C1) was suppressed by dexamethasone at 48 h, suggesting limited effect of the drug in prolonged LSEC culture. Conclusion: The study presents a detailed overview of biological processes and pathways affected by dexamethasone in mouse LSECs in vitro.
Collapse
|
28
|
Özgüldez HÖ, Bulut-Karslioğlu A. Dormancy, Quiescence, and Diapause: Savings Accounts for Life. Annu Rev Cell Dev Biol 2024; 40:25-49. [PMID: 38985838 DOI: 10.1146/annurev-cellbio-112122-022528] [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] [Indexed: 07/12/2024]
Abstract
Life on Earth has been through numerous challenges over eons and, one way or another, has always triumphed. From mass extinctions to more daily plights to find food, unpredictability is everywhere. The adaptability of life-forms to ever-changing environments is the key that confers life's robustness. Adaptability has become synonymous with Darwinian evolution mediated by heritable genetic changes. The extreme gene-centric view, while being of central significance, at times has clouded our appreciation of the cell as a self-regulating entity informed of, and informing, the genetic data. An essential element that powers adaptability is the ability to regulate cell growth. In this review, we provide an extensive overview of growth regulation spanning species, tissues, and regulatory mechanisms. We aim to highlight the commonalities, as well as differences, of these phenomena and their molecular regulators. Finally, we curate open questions and areas for further exploration.
Collapse
Affiliation(s)
- Hatice Özge Özgüldez
- Stem Cell Chromatin Group, Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany;
| | - Aydan Bulut-Karslioğlu
- Stem Cell Chromatin Group, Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany;
| |
Collapse
|
29
|
Tolue Ghasaban F, Taghehchian N, Zangouei AS, Keivany MR, Moghbeli M. MicroRNA-135b mainly functions as an oncogene during tumor progression. Pathol Res Pract 2024; 262:155547. [PMID: 39151250 DOI: 10.1016/j.prp.2024.155547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
Late diagnosis is considered one of the main reasons of high mortality rate among cancer patients that results in therapeutic failure and tumor relapse. Therefore, it is needed to evaluate the molecular mechanisms associated with tumor progression to introduce efficient markers for the early tumor detection among cancer patients. The remarkable stability of microRNAs (miRNAs) in body fluids makes them potential candidates to use as the non-invasive tumor biomarkers in cancer screening programs. MiR-135b has key roles in prognosis and survival of cancer patients by either stimulating or inhibiting cell proliferation, invasion, and angiogenesis. Therefore, in the present review we assessed the molecular biology of miR-135b during tumor progression to introduce that as a novel tumor marker in cancer patients. It has been reported that miR-135b mainly acts as an oncogene by regulation of transcription factors, signaling pathways, drug response, cellular metabolism, and autophagy. This review paves the way to suggest miR-135b as a tumor marker and therapeutic target in cancer patients following the further clinical trials and animal studies.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keivany
- Department of Radiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Santini L, Kowald S, Cerron-Alvan LM, Huth M, Fabing AP, Sestini G, Rivron N, Leeb M. FoxO transcription factors actuate the formative pluripotency specific gene expression programme. Nat Commun 2024; 15:7879. [PMID: 39251582 PMCID: PMC11384738 DOI: 10.1038/s41467-024-51794-9] [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: 01/24/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Naïve pluripotency is sustained by a self-reinforcing gene regulatory network (GRN) comprising core and naïve pluripotency-specific transcription factors (TFs). Upon exiting naïve pluripotency, embryonic stem cells (ESCs) transition through a formative post-implantation-like pluripotent state, where they acquire competence for lineage choice. However, the mechanisms underlying disengagement from the naïve GRN and initiation of the formative GRN are unclear. Here, we demonstrate that phosphorylated AKT acts as a gatekeeper that prevents nuclear localisation of FoxO TFs in naïve ESCs. PTEN-mediated reduction of AKT activity upon exit from naïve pluripotency allows nuclear entry of FoxO TFs, enforcing a cell fate transition by binding and activating formative pluripotency-specific enhancers. Indeed, FoxO TFs are necessary and sufficient for the activation of the formative pluripotency-specific GRN. Our work uncovers a pivotal role for FoxO TFs in establishing formative post-implantation pluripotency, a critical early embryonic cell fate transition.
Collapse
Affiliation(s)
- Laura Santini
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Saskia Kowald
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Luis Miguel Cerron-Alvan
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Michelle Huth
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Anna Philina Fabing
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Giovanni Sestini
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Nicolas Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria.
| |
Collapse
|
31
|
Grant MP, Alatassi R, Diab MO, Abushal M, Epure LM, Huk OL, Bergeron SG, Im Sampen HJ, Antoniou J, Mwale F. Cobalt ions induce a cellular senescence secretory phenotype in human synovial fibroblast-like cells that may be an early event in the development of adverse local tissue reactions to hip implants. OSTEOARTHRITIS AND CARTILAGE OPEN 2024; 6:100490. [PMID: 38828014 PMCID: PMC11141261 DOI: 10.1016/j.ocarto.2024.100490] [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: 07/03/2023] [Accepted: 05/09/2024] [Indexed: 06/05/2024] Open
Abstract
Objectives Total hip arthroplasty is a successful procedure for treating advanced osteoarthritis (OA). Metal bearing surfaces remain one of the most widely implanted prosthesis, however approximately 10% of patients develop adverse local tissue reactions (ALTRs), namely lymphocytic predominant soft tissue reaction with or without necrosis and osteolysis resulting in high revision rates. The mechanism(s) for these reactions remains unclear although T lymphocyte mediated type IV hypersensitivity to cobalt (Co) and chromium (Cr) ions have been described. The purpose of this study was to determine the prolonged effects of Co and Cr metal ions on synovial fibroblasts to better understand the impact of the synovial membrane in the development of ALTRs. Methods Human synovial fibroblast-like cells were isolated from donors undergoing arthroplasty. DNA content and Alamar blue assay were used to determine cellular viability against exposure to Co and Cr. A beta-galactosidase assay was used to determine the development of cellular senescence. Western blotting and RT-qPCR were employed to determine changes in senescent associated secretory factors, signaling and anti-oxidant enzyme expression. A fluorescent assay was used to measure accumulation of hydrogen peroxide. Results We demonstrate that prolonged cobalt exposure results in a downregulation of the enzyme catalase resulting in cytosolic accumulation of hydrogen peroxide, decreased Akt activity and cellular senescence. Senescent fibroblasts demonstrated upregulation of proinflammatory cytokines IL-1β and TNFα in addition to the neurotrophic factor NGF. Conclusion Our results provide evidence that metal ions induce a senescent associated secretory phenotype in synovial fibroblasts that could contribute to the development of adverse local tissue reactions.
Collapse
Affiliation(s)
- Michael P. Grant
- Department of Surgery, McGill University, Montreal, Canada
- SMBD-Jewish General Hospital, Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Road, Room F-602, Montreal, Quebec, H3T 1E2, Canada
| | | | | | | | - Laura M. Epure
- Department of Surgery, McGill University, Montreal, Canada
| | - Olga L. Huk
- Department of Surgery, McGill University, Montreal, Canada
- SMBD-Jewish General Hospital, Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Road, Room F-602, Montreal, Quebec, H3T 1E2, Canada
| | - Stephane G. Bergeron
- Department of Surgery, McGill University, Montreal, Canada
- SMBD-Jewish General Hospital, Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Road, Room F-602, Montreal, Quebec, H3T 1E2, Canada
| | - Hee-Jeong Im Sampen
- Department of Biomedical Engineering, University of Illinois Chicago, IL, USA
| | - John Antoniou
- Department of Surgery, McGill University, Montreal, Canada
- SMBD-Jewish General Hospital, Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Road, Room F-602, Montreal, Quebec, H3T 1E2, Canada
| | - Fackson Mwale
- Department of Surgery, McGill University, Montreal, Canada
- SMBD-Jewish General Hospital, Lady Davis Institute for Medical Research, 3755 Cote Ste-Catherine Road, Room F-602, Montreal, Quebec, H3T 1E2, Canada
| |
Collapse
|
32
|
Menges CW, Hassan D, Cheung M, Bellacosa A, Testa JR. Alterations of the AKT Pathway in Sporadic Human Tumors, Inherited Susceptibility to Cancer, and Overgrowth Syndromes. Curr Top Microbiol Immunol 2024. [PMID: 39192048 DOI: 10.1007/82_2024_278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The AKT kinases are critical signaling molecules that regulate cellular physiology upon the activation of tyrosine kinase receptors and phosphatidylinositol 3-kinases (PI3K). AKT kinases govern many cellular processes considered hallmarks of cancer, including cell proliferation and survival, cell size, tumor invasion, metastasis, and angiogenesis. AKT signaling is regulated by multiple tumor suppressors and oncogenic proteins whose loss or activation, respectively, leads to dysregulation of this pathway, thereby contributing to oncogenesis. Herein, we review the enormous body of literature documenting how the AKT pathway becomes hyperactivated in sporadic human tumors and various hereditary cancer syndromes. We also discuss the role of activating mutations of AKT pathway genes in various chimeric overgrowth disorders, including Proteus syndrome, hypoglycemia with hypertrophy, CLOVES and SOLAMEN syndromes, and hemimegalencephaly.
Collapse
Affiliation(s)
- Craig W Menges
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Eurofins Lancaster Laboratories Professional Scientific Services, Lancaster, PA, 17601, USA
| | - Dalal Hassan
- Cancer Epigenetics Institute, Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
- Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mitchell Cheung
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Alfonso Bellacosa
- Cancer Epigenetics Institute, Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Joseph R Testa
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| |
Collapse
|
33
|
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: 0] [Impact Index Per Article: 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
|
34
|
Gao C, Nie H. Exploring the Heat-Responsive miRNAs and their Target Gene Regulation in Ruditapes philippinarum Under Acute Heat Stress. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:810-826. [PMID: 39046591 DOI: 10.1007/s10126-024-10348-2] [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: 04/30/2024] [Accepted: 07/13/2024] [Indexed: 07/25/2024]
Abstract
This study aimed to investigate the inherent molecular regulatory mechanisms of Ruditapes philippinarum in response to extremely high-temperature environments and to enhance the sustainable development of the R. philippinarum aquaculture industry. In this study, we established a differential expression profile of miRNA under acute heat stress and identified a total of 46 known miRNAs and 80 novel miRNAs, three of which were detected to be significantly differentially expressed. We analyzed the functions of target genes regulated by differentially expressed miRNAs (DEMs) of R. philippinarum. The findings of the KEGG enrichment analysis revealed that 29 enriched pathways in the group were subjected to acute heat stress. Notably, fatty acid metabolism, FoxO signaling pathway, TGF-β signaling pathway, and ubiquitin-mediated proteolysis were found to play significant roles in response to acute heat stress. We established a regulatory map of DEMs and their target genes in response to heat stress and constructed the miRNA-mRNA regulation network. This study provides valuable insights into the response of R. philippinarum to high temperature, helping to understand its underlying molecular regulatory mechanisms under high-temperature stress.
Collapse
Affiliation(s)
- Changsheng Gao
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, Dalian Ocean University, Dalian, 116023, China
| | - Hongtao Nie
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, 116023, China.
- Engineering and Technology Research Center of Shellfish Breeding in Liaoning Province, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
35
|
Sato R, Vatic M, Peixoto da Fonseca GW, Anker SD, von Haehling S. Biological basis and treatment of frailty and sarcopenia. Cardiovasc Res 2024:cvae073. [PMID: 38828887 DOI: 10.1093/cvr/cvae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/23/2022] [Accepted: 12/20/2022] [Indexed: 06/05/2024] Open
Abstract
In an ageing society, the importance of maintaining healthy life expectancy has been emphasized. As a result of age-related decline in functional reserve, frailty is a state of increased vulnerability and susceptibility to adverse health outcomes with a serious impact on healthy life expectancy. The decline in skeletal muscle mass and function, also known as sarcopenia, is key in the development of physical frailty. Both frailty and sarcopenia are highly prevalent in patients not only with advanced age but also in patients with illnesses that exacerbate their progression like heart failure (HF), cancer, or dementia, with the prevalence of frailty and sarcopenia in HF patients reaching up to 50-75% and 19.5-47.3%, respectively, resulting in 1.5-3 times higher 1-year mortality. The biological mechanisms of frailty and sarcopenia are multifactorial, complex, and not yet fully elucidated, ranging from DNA damage, proteostasis impairment, and epigenetic changes to mitochondrial dysfunction, cellular senescence, and environmental factors, many of which are further linked to cardiac disease. Currently, there is no gold standard for the treatment of frailty and sarcopenia, however, growing evidence supports that a combination of exercise training and nutritional supplement improves skeletal muscle function and frailty, with a variety of other therapies being devised based on the underlying pathophysiology. In this review, we address the involvement of frailty and sarcopenia in cardiac disease and describe the latest insights into their biological mechanisms as well as the potential for intervention through exercise, diet, and specific therapies.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité; German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
36
|
Carmona-Aldana F, Yong LW, Reinberg D, Desplan C. Phenomenon of reproductive plasticity in ants. CURRENT OPINION IN INSECT SCIENCE 2024; 63:101197. [PMID: 38583769 PMCID: PMC11139587 DOI: 10.1016/j.cois.2024.101197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/26/2024] [Accepted: 03/31/2024] [Indexed: 04/09/2024]
Abstract
Ant colonies are organized in castes with distinct behaviors that together allow the colony to strive. Reproduction relies on one or a few queens that stay in the nest producing eggs, while females of the worker caste do not reproduce and instead engage in colony maintenance and brood caretaking. Yet, in spite of this clear separation of functions, workers can become reproductive under defined circumstances. Here, we review the context in which workers become reproductive, exhibiting asexual or sexual reproduction depending on the species. Remarkably, the activation of reproduction in these workers can be quite stable, with changes that include behavior and a dramatic extension of lifespan. We compare these changes between species that do or do not have a queen caste. We discuss how the mechanisms underlying reproductive plasticity include changes in hormonal functions and in epigenetic configurations. Further studies are warranted to elucidate not only how reproductive functions have been gradually restricted to the queen caste during evolution but also how reproductive plasticity remains possible in workers of some species.
Collapse
Affiliation(s)
| | - Luok Wen Yong
- Department of Biology, New York University, NY 10003, USA
| | - Danny Reinberg
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Coral Gables, FL 33124, USA.
| | - Claude Desplan
- Department of Biology, New York University, NY 10003, USA; Center for Genomics and Systems Biology, New York University, Abu Dhabi 51133, United Arab Emirates.
| |
Collapse
|
37
|
de Brot S, Cobb J, Alibhai AA, Jackson-Oxley J, Haque M, Patke R, Harris AE, Woodcock CL, Lothion-Roy J, Varun D, Thompson R, Gomes C, Kubale V, Dunning MD, Jeyapalan JN, Mongan NP, Rutland CS. Immunohistochemical Investigation into Protein Expression Patterns of FOXO4, IRF8 and LEF1 in Canine Osteosarcoma. Cancers (Basel) 2024; 16:1945. [PMID: 38792023 PMCID: PMC11120020 DOI: 10.3390/cancers16101945] [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: 04/30/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
Osteosarcoma (OSA) is the most common type of primary bone malignancy in people and dogs. Our previous molecular comparisons of canine OSA against healthy bone resulted in the identification of differentially expressed protein-expressing genes (forkhead box protein O4 (FOXO4), interferon regulatory factor 8 (IRF8), and lymphoid enhancer binding factor 1 (LEF1)). Immunohistochemistry (IHC) and H-scoring provided semi-quantitative assessment of nuclear and cytoplasmic staining alongside qualitative data to contextualise staining (n = 26 patients). FOXO4 was expressed predominantly in the cytoplasm with significantly lower nuclear H-scores. IRF8 H-scores ranged from 0 to 3 throughout the cohort in the nucleus and cytoplasm. LEF1 was expressed in all patients with significantly lower cytoplasmic staining compared to nuclear. No sex or anatomical location differences were observed. While reduced levels of FOXO4 might indicate malignancy, the weak or absent protein expression limits its primary use as diagnostic tumour marker. IRF8 and LEF1 have more potential for prognostic and diagnostic uses and facilitate further understanding of their roles within their respective molecular pathways, including Wnt/beta-catenin/LEF1 signalling and differential regulation of tumour suppressor genes. Deeper understanding of the mechanisms involved in OSA are essential contributions towards the development of novel diagnostic, prognostic, and treatment options in human and veterinary medicine contexts.
Collapse
Affiliation(s)
- Simone de Brot
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
- Comparative Pathology Platform of the University of Bern (COMPATH), Institute of Animal Pathology, University of Bern, 3012 Bern, Switzerland
| | - Jack Cobb
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Aziza A. Alibhai
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Jorja Jackson-Oxley
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Maria Haque
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Rodhan Patke
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Anna E. Harris
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Corinne L. Woodcock
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Jennifer Lothion-Roy
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Dhruvika Varun
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Rachel Thompson
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Claudia Gomes
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
| | - Valentina Kubale
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Mark D. Dunning
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
- Willows Veterinary Centre and Referral Service, Solihull B90 4NH, UK
| | - Jennie N. Jeyapalan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
- Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nigel P. Mongan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
- Willows Veterinary Centre and Referral Service, Solihull B90 4NH, UK
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10075, USA
| | - Catrin S. Rutland
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK; (S.d.B.); (J.C.); (A.A.A.); (J.J.-O.); (M.H.); (R.P.); (A.E.H.); (C.L.W.); (J.L.-R.); (D.V.); (R.T.); (C.G.); (M.D.D.); (J.N.J.)
- Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
38
|
Gupta A, Haldhar R, Agarwal V, Rajput DS, Chun KS, Han SB, Raj V, Lee S. Exploring the Potential of Natural Products as FoxO1 Inhibitors: an In Silico Approach. Biomol Ther (Seoul) 2024; 32:390-398. [PMID: 38586882 PMCID: PMC11063485 DOI: 10.4062/biomolther.2023.156] [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: 09/05/2023] [Revised: 10/14/2023] [Accepted: 10/21/2023] [Indexed: 04/09/2024] Open
Abstract
FoxO1, a member of the Forkhead transcription factor family subgroup O (FoxO), is expressed in a range of cell types and is crucial for various pathophysiological processes, such as apoptosis and inflammation. While FoxO1's roles in multiple diseases have been recognized, the target has remained largely unexplored due to the absence of cost-effective and efficient inhibitors. Therefore, there is a need for natural FoxO1 inhibitors with minimal adverse effects. In this study, docking, MMGBSA, and ADMET analyses were performed to identify natural compounds that exhibit strong binding affinity to FoxO1. The top candidates were then subjected to molecular dynamics (MD) simulations. A natural product library was screened for interaction with FoxO1 (PDB ID- 3CO6) using the Glide module of the Schrödinger suite. In silico ADMET profiling was conducted using SwissADME and pkCSM web servers. Binding free energies of the selected compounds were assessed with the Prime-MMGBSA module, while the dynamics of the top hits were analyzed using the Desmond module of the Schrödinger suite. Several natural products demonstrated high docking scores with FoxO1, indicating their potential as FoxO1 inhibitors. Specifically, the docking scores of neochlorogenic acid and fraxin were both below -6.0. These compounds also exhibit favorable drug-like properties, and a 25 ns MD study revealed a stable interaction between fraxin and FoxO1. Our findings highlight the potential of various natural products, particularly fraxin, as effective FoxO1 inhibitors with strong binding affinity, dynamic stability, and suitable ADMET profiles.
Collapse
Affiliation(s)
- Anugya Gupta
- Faculty of Medical and Paramedical Sciences, Madhyanchal Professional University, Bhopal 462044, Madhya Pradesh, India
| | - Rajesh Haldhar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, Uttar Pradesh, India
| | - Dharmendra Singh Rajput
- Faculty of Medical and Paramedical Sciences, Madhyanchal Professional University, Bhopal 462044, Madhya Pradesh, India
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Sang Beom Han
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Vinit Raj
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
39
|
Zhao L, Xue H, Elumalai P, Zhu X, Wang L, Zhang K, Li D, Ji J, Luo J, Cui J, Gao X. Sublethal acetamiprid affects reproduction, development and disrupts gene expression in Binodoxys communis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024:10.1007/s11356-024-33415-6. [PMID: 38656721 DOI: 10.1007/s11356-024-33415-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
At present, understanding of neonicotinoid toxicity in arthropods remains limited. We here evaluated the lethal and sublethal effects of acetamiprid in F0 and F1 generations of Binodoxys communis using a range of sublethal concentrations. The 10% lethal concentration (LC10) and half lethal concentration (LC25) of ACE had negative effects on the B. communis survival rate, adult longevity, parasitism rate, and emergence rate, and significantly prolonged the duration of the developmental cycle. ACE also had intergenerational effects, with some biological indices affected in the F1 generation after pesticide exposure. Transcriptomic analysis demonstrated that differentially expressed genes were enriched in specific pathways including the amino acid metabolism, carbohydrate metabolism, energy metabolism, exogenous metabolism, signal transduction, and glutathione metabolism pathways. These results indicated strong contact toxicity of ACE to B. communis, which may inhibit their biological control capacity. These results improve our understanding of the toxicological mechanisms of parasitic natural enemies in response to insecticide exposure.
Collapse
Affiliation(s)
- Likang Zhao
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hui Xue
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Punniyakotti Elumalai
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiangzhen Zhu
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Li Wang
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Kaixin Zhang
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Dongyang Li
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jichao Ji
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Junyu Luo
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jinjie Cui
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xueke Gao
- Zhengzhou Research Base, State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, Henan, China.
- State Key Laboratory of Cotton Bio-breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
40
|
Sekar AP, Nurmala S, Matsuura E, Tan XW, Rahmasari R, Sauriasari R. Estrogen Receptor Is Required for Metformin-Induced Apoptosis in Breast Cancer Cells Under Hyperglycemic Conditions. Breast Cancer (Auckl) 2024; 18:11782234241240173. [PMID: 38779416 PMCID: PMC11110926 DOI: 10.1177/11782234241240173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/29/2024] [Indexed: 05/25/2024] Open
Abstract
Backgrounds About 25% to 30% of estrogen receptor (ER)-positive breast cancer patients develop resistance to endocrine therapy. Human epidermal growth factor receptor 2 (HER2) has been shown to cooperate with several growth factors that regulate cellular energy metabolism, including the insulin-like growth factor 1 receptor (IGF-1R). Objective As the first-line therapy for type 2 diabetes mellitus (T2DM) patients, metformin is widely known to inhibit the metabolic reprogramming of cancer cells. This study aims to investigate metformin's efficacy in inhibiting endocrine resistance related to genes regulating energy metabolism in both ER-positive and ER-negative breast cancer cell lines under hyperglycemic conditions. Design and methods MDA-MB-361 (ER-positive, HER2-positive) and SKBR3 (ER-negative, HER2-positive) cancer cell lines were used to represent ER status. Cell viability and cell survival rate were measured using the colorimetric assay of Cell Counting Kit-8. All mRNA levels were quantified using real-time quantitative polymerase chain reaction preceded by reverse transcription. A P value of <.05 was considered statistically significant. Results Unlike MDA-MB-361, SKBR3 were found to acquire resistance upon metformin treatment in hyperglycemic conditions. Moreover, the mRNA expression of IGF-1R and its downstream signaling, such as the mammalian target of rapamycin (mTOR), was not affected by metformin. Meanwhile, the mRNA expression level of ribosomal S6 kinase 1 (S6K1) was upregulated, whereas forkhead box O1 (FOXO1) was downregulated after metformin treatment in hyperglycemic conditions. Conclusions This preliminary study suggests that an alternative pathway of metformin resistance may exist in the absence of ERα. Therefore, relying solely on metformin may be inadequate to inhibit the aggressiveness of breast cancer cells.
Collapse
Affiliation(s)
- Andisyah Putri Sekar
- Pharmacology and Clinical Pharmacy Laboratory, Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| | - Septia Nurmala
- Pharmacology and Clinical Pharmacy Laboratory, Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| | - Eiji Matsuura
- Graduate School of Interdiscplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Xian Wen Tan
- Collaborative Research Center for Okayama Medical Innovation Center & Department of Cell Chemistry, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ratika Rahmasari
- Microbiology and Biotechnology Laboratory, Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| | - Rani Sauriasari
- Pharmacology and Clinical Pharmacy Laboratory, Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia
| |
Collapse
|
41
|
Abugessaisa I, Manabe RI, Kawashima T, Tagami M, Takahashi C, Okazaki Y, Bandinelli S, Kasukawa T, Ferrucci L. OVCH1 Antisense RNA 1 is differentially expressed between non-frail and frail old adults. GeroScience 2024; 46:2063-2081. [PMID: 37817005 PMCID: PMC10828349 DOI: 10.1007/s11357-023-00961-9] [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: 06/23/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
While some old adults stay healthy and non-frail up to late in life, others experience multimorbidity and frailty often accompanied by a pro-inflammatory state. The underlying molecular mechanisms for those differences are still obscure. Here, we used gene expression analysis to understand the molecular underpinning between non-frail and frail individuals in old age. Twenty-four adults (50% non-frail and 50% frail) from InCHIANTI study were included. Total RNA extracted from whole blood was analyzed by Cap Analysis of Gene Expression (CAGE). CAGE identified transcription start site (TSS) and active enhancer regions. We identified a set of differentially expressed (DE) TSS and enhancer between non-frail and frail and male and female participants. Several DE TSSs were annotated as lncRNA (XIST and TTTY14) and antisense RNAs (ZFX-AS1 and OVCH1 Antisense RNA 1). The promoter region chr6:366,786,54-366,787,97;+ was DE and overlapping the longevity CDKN1A gene. GWAS-LD enrichment analysis identifies overlapping LD-blocks with the DE regions with reported traits in GWAS catalog (isovolumetric relaxation time and urinary tract infection frequency). Furthermore, we used weighted gene co-expression network analysis (WGCNA) to identify changes of gene expression associated with clinical traits and identify key gene modules. We performed functional enrichment analysis of the gene modules with significant trait/module correlation. One gene module is showing a very distinct pattern in hub genes. Glycogen Phosphorylase L (PYGL) was the top ranked hub gene between non-frail and frail. We predicted transcription factor binding sites (TFBS) and motif activity. TF involved in age-related pathways (e.g., FOXO3 and MYC) shows different expression patterns between non-frail and frail participants. Expanding the study of OVCH1 Antisense RNA 1 and PYGL may help understand the mechanisms leading to loss of homeostasis that ultimately causes frailty.
Collapse
Affiliation(s)
- Imad Abugessaisa
- Laboratory for Large-Scale Biomedical Data Technology, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan.
| | - Ri-Ichiroh Manabe
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Tsugumi Kawashima
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Michihira Tagami
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Chitose Takahashi
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Yasushi Okazaki
- Laboratory for Comprehensive Genomic Analysis, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Stefania Bandinelli
- Azienda USL Toscana Centro, InCHIANTI, Villa Margherita, Primo piano Viale Michelangelo, 41, 50125, Firenze, Italy
| | - Takeya Kasukawa
- Laboratory for Large-Scale Biomedical Data Technology, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
- Institute for Protein Research, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital 5th floor, 3001 S. Hanover Street, Baltimore, MD, 21225, USA
| |
Collapse
|
42
|
Alruhaimi RS, Hassanein EHM, Bin-Jumah MN, Mahmoud AM. Cadmium-induced lung injury is associated with oxidative stress, apoptosis, and altered SIRT1 and Nrf2/HO-1 signaling; protective role of the melatonin agonist agomelatine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2335-2345. [PMID: 37819390 DOI: 10.1007/s00210-023-02754-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Cadmium (Cd) is a hazardous heavy metal extensively employed in manufacturing polyvinyl chloride, batteries, and other industries. Acute lung injury has been directly connected to Cd exposure. Agomelatine (AGM), a melatonin analog, is a drug licensed for treating severe depression. This study evaluated the effect of AGM against Cd-induced lung injury in rats. AGM was administered in a dose of 25 mg/kg/day orally, while cadmium chloride (CdCl2) was injected intraperitoneally in a dose of 1.2 mg/kg to induce lung injury. Pre-treatment with AGM remarkably ameliorated Cd-induced lung histopathological abrasions. AGM decreased reactive oxygen species (ROS) production, lipid peroxidation, suppressed NDAPH oxidase, and boosted the antioxidants. AGM increased Nrf2, GCLC, HO-1, and TNXRD1 mRNA, as well as HO-1 activity and downregulated Keap1. AGM downregulated Bax and caspase-3 and upregulated Bcl-2, SIRT1, and FOXO3 expression levels in the lung. In conclusion, AGM has a protective effect against Cd-induced lung injury via its antioxidant and anti-apoptotic effects mediated via regulating Nrf2/HO-1 and SIRT1/FOXO3 signaling.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71562, Egypt
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Ayman M Mahmoud
- Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK.
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
43
|
Yan J, Zhou L, Liu M, Zhu H, Zhang X, Cai E, Xu X, Chen T, Cheng H, Liu J, Wang S, Dai L, Chang X, Tang F. Single-cell analysis reveals insights into epithelial abnormalities in ovarian endometriosis. Cell Rep 2024; 43:113716. [PMID: 38412094 DOI: 10.1016/j.celrep.2024.113716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/19/2023] [Accepted: 01/11/2024] [Indexed: 02/29/2024] Open
Abstract
Ovarian endometriosis is characterized by the growth of endometrial tissue within the ovary, causing infertility and chronic pain. However, its pathophysiology remains unclear. Utilizing high-precision single-cell RNA sequencing, we profile the normal, eutopic, and ectopic endometrium from 34 individuals across proliferative and secretory phases. We observe an increased proportion of ciliated cells in both eutopic and ectopic endometrium, characterized by a diminished expression of estrogen sulfotransferase, which likely confers apoptosis resistance. After translocating to ectopic lesions, endometrial epithelium upregulates nicotinamide N-methyltransferase expression that inhibits apoptosis by promoting deacetylation and subsequent nuclear exclusion of transcription factor forkhead box protein O1, thereby leading to the downregulation of the apoptotic gene BIM. Moreover, epithelial cells in ectopic lesions elevate HLA class II complex expression, which stimulates CD4+ T cells and consequently contributes to chronic inflammation. Altogether, our study provides a comprehensive atlas of ovarian endometriosis and highlights potential therapeutic targets for modulating apoptosis and inflammation.
Collapse
Affiliation(s)
- Jia Yan
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Mengya Liu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Honglan Zhu
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Xin Zhang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China
| | - E Cai
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Xueqiang Xu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Tinghan Chen
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Jun'e Liu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Shang Wang
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Lin Dai
- Department of Pathology, People's Hospital, Peking University, Beijing 100044, China
| | - Xiaohong Chang
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China.
| | - Fuchou Tang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China.
| |
Collapse
|
44
|
Torigoe TH, Willcox DC, Shimabukuro M, Higa M, Gerschenson M, Andrukhiv A, Suzuki M, Morris BJ, Chen R, Gojanovich GS, Allsopp RC, Willcox BJ. Novel protective effect of the FOXO3 longevity genotype on mechanisms of cellular aging in Okinawans. NPJ AGING 2024; 10:18. [PMID: 38459055 PMCID: PMC10923797 DOI: 10.1038/s41514-024-00142-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/06/2024] [Indexed: 03/10/2024]
Abstract
The genetic association of FOXO3 genotypes with human longevity is well established, although the mechanism is not fully understood. We now report on the relationship of the FOXO3 longevity variant rs2802292 with telomere length, telomerase activity, FOXO3 expression, and inflammatory cytokine levels in men and women. In agreement with earlier work, the FOXO3 longevity variant conferred protection against telomere shortening of peripheral blood mononuclear cells from adults aged 55 years and older. This was accompanied by higher levels of telomerase activity in mononuclear cells for carriers of the longevity-associated FOXO3 G-allele of SNP rs2802292 (P = 0.015). FOXO3 mRNA expression increased slightly with age in both young (P = 0.02) and old (P = 0.08) G-allele carriers. Older female G-allele carriers displayed a modest decline in levels of pro-inflammatory cytokine IL-6 with age (P = 0.07). In contrast, older male G-allele carriers displayed an age-dependent increase in levels of anti-inflammatory cytokine IL-10 with age (P = 0.04). Thus, FOXO3 may act through several different pro-longevity mechanisms, which may differ by age and sex.
Collapse
Affiliation(s)
- Trevor H Torigoe
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| | - D Craig Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Department of Human Welfare, Okinawa International University, Ginowan, Okinawa, Japan
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| | - Michio Shimabukuro
- Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University School of Medicine, Fukushima, Fukushima, Japan
- Diabetes and Life-Style Related Disease Center, Tomishiro Central Hospital, Tomishiro, Okinawa, Japan
| | - Moritake Higa
- Diabetes and Life-Style Related Disease Center, Tomishiro Central Hospital, Tomishiro, Okinawa, Japan
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Anastasia Andrukhiv
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Makoto Suzuki
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
| | - Brian J Morris
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Randi Chen
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| | - Greg S Gojanovich
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Richard C Allsopp
- Institute for Biogenesis Research, Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan.
| | - Bradley J Willcox
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
- Okinawa Research Center for Longevity Science, Urasoe, Okinawa, Japan
- Department of Research, Kuakini Medical Center, Honolulu, HI, USA
| |
Collapse
|
45
|
Shamnas v M, Singh A, Kumar A, Mishra GP, Sinha SK. Exitrons: offering new roles to retained introns-the novel regulators of protein diversity and utility. AOB PLANTS 2024; 16:plae014. [PMID: 38566894 PMCID: PMC10985678 DOI: 10.1093/aobpla/plae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Exitrons are exonic introns. This subclass of intron retention alternative splicing does not contain a Pre-Terminating stop Codon. Therefore, when retained, they are always a part of a protein. Intron retention is a frequent phenomenon predominantly found in plants, which results in either the degradation of the transcripts or can serve as a stable intermediate to be processed upon induction by specific signals or the cell status. Interestingly, exitrons have coding ability and may confer additional attributes to the proteins that retain them. Therefore, exitron-containing and exitron-spliced isoforms will be a driving force for creating protein diversity in the proteome of an organism. This review establishes a basic understanding of exitron, discussing its genesis, key features, identification methods and functions. We also try to depict its other potential roles. The present review also aims to provide a fundamental background to those who found such exitronic sequences in their gene(s) and to speculate the future course of studies.
Collapse
Affiliation(s)
- Muhammed Shamnas v
- ICAR-National Institute for Plant Biotechnology, Pusa Campus, New Delhi 110012, India
| | - Akanksha Singh
- ICAR-National Institute for Plant Biotechnology, Pusa Campus, New Delhi 110012, India
- Department of Botany and Plant Pathology, Lilly Hall of Life Sciences, Purdue University, West Lafayette 47906, Indiana, USA
| | - Anuj Kumar
- ICAR-National Institute for Plant Biotechnology, Pusa Campus, New Delhi 110012, India
| | - Gyan Prakash Mishra
- Division of Genetics, ICAR-Indian Agricultural Research Institute, Pusa Campus, New Delhi 110012, India
| | - Subodh Kumar Sinha
- ICAR-National Institute for Plant Biotechnology, Pusa Campus, New Delhi 110012, India
| |
Collapse
|
46
|
Chen L, Liang J, Zhang Q, Yang C, Lu H, Zhang R, Chen K, Wang S, Li M, Zhang S, He N. Mulberry-derived miR168a downregulates BmMthl1 to promote physical development and fecundity in silkworms. Int J Biol Macromol 2024; 259:129077. [PMID: 38199542 DOI: 10.1016/j.ijbiomac.2023.129077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Plant-derived miRNAs and their interactions with host organisms are considered important factors in regulating host physiological processes. In this study, we investigated the interaction between the silkworm, an oligophagous insect, and its primary food source, mulberry, to determine whether mulberry-derived miRNAs can penetrate silkworm cells and regulate their functions. Our results demonstrated that miR168a from mulberry leaves enters the silkworm hemolymph and binds to the silkworm Argonaute1 BmAGO1, which is transported via vesicles secreted by silkworm cells to exert its regulatory functions. In vivo and in vitro functional studies revealed that miR168a targets the mRNA of silkworm G protein-coupled receptor, BmMthl1, thereby inhibiting its expression and activating the JNK-FoxO pathway. This activation reduces oxidative stress responses, prolongs the lifespan of silkworms, and improves their reproductive capacity. These findings highlight the challenges of replacing mulberry leaves with alternative protein sources and provide a foundation for developing silkworm germplasms suitable for factory rearing.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Jiubo Liang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Qi Zhang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Chao Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Hulin Lu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Renze Zhang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Kaiying Chen
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Sheng Wang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Mingbo Li
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Shaoyu Zhang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Ningjia He
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China.
| |
Collapse
|
47
|
Yu J, Chen G, Zhu H, Zhong Y, Yang Z, Jian Z, Xiong X. Metabolic and proteostatic differences in quiescent and active neural stem cells. Neural Regen Res 2024; 19:43-48. [PMID: 37488842 PMCID: PMC10479840 DOI: 10.4103/1673-5374.375306] [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: 09/20/2022] [Revised: 02/16/2023] [Accepted: 04/17/2023] [Indexed: 07/26/2023] Open
Abstract
Adult neural stem cells are neurogenesis progenitor cells that play an important role in neurogenesis. Therefore, neural regeneration may be a promising target for treatment of many neurological illnesses. The regenerative capacity of adult neural stem cells can be characterized by two states: quiescent and active. Quiescent adult neural stem cells are more stable and guarantee the quantity and quality of the adult neural stem cell pool. Active adult neural stem cells are characterized by rapid proliferation and differentiation into neurons which allow for integration into neural circuits. This review focuses on differences between quiescent and active adult neural stem cells in nutrition metabolism and protein homeostasis. Furthermore, we discuss the physiological significance and underlying advantages of these differences. Due to the limited number of adult neural stem cells studies, we referred to studies of embryonic adult neural stem cells or non-mammalian adult neural stem cells to evaluate specific mechanisms.
Collapse
Affiliation(s)
- Jiacheng Yu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Gang Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhenxing Yang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
48
|
Xu Q, Dong S, Gong Q, Dai Q, Cheng R, Ge Y. The Tanshinones (Tan) Extract From Salvia miltiorrhiza Bunge Induces ROS-Dependent Apoptosis in Pancreatic Cancer via AKT Hyperactivation-Mediated FOXO3/SOD2 Signaling. Integr Cancer Ther 2024; 23:15347354241258961. [PMID: 38899834 PMCID: PMC11191618 DOI: 10.1177/15347354241258961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024] Open
Abstract
CONTEXT Salvia miltiorrhiza (SM) is a commonly used herb in traditional Chinese medicine (TCM) and has been used in the treatment of pancreatic cancer to relieve the symptom of "blood stasis and toxin accumulation." Tanshinones (Tan), the main lipophilic constituents extracted from the roots and rhizomes of SM, have been reported to possess anticancer functions in several cancers. But the mechanism of how the active components work in pancreatic cancer still need to be clarified. OBJECTIVE In this study, we aimed to investigate the therapeutic potential of Tan in pancreatic cancer and elucidate the underlying mechanisms. MATERIALS AND METHODS The viabilities of PANC-1 and Bxpc-3 cells were determined by MTT assay, after treatment with various concentrations of Tan. The apoptotic cells were quantified by annexin V-FITC/PI staining and DAPI staining assays. The expression of relative proteins was used western blotting. Tumor growth was assessed by subcutaneously inoculating cells into C57BL/6 mice. RESULTS Our experiments discovered that Tan effectively suppressed pancreatic cancer cell proliferation and promoted apoptosis. Mechanistically, we propose that Tan enhances intracellular ROS levels by activating the AKT/FOXO3/SOD2 signaling pathway, ultimately leading to apoptosis in pancreatic cancer cells. In vivo assay showed the antitumor effect of Tan. CONCLUSION Tan, a natural compound from Salvia miltiorrhiza, was found to effectively suppress pancreatic cancer cell proliferation and promote apoptosis both in vitro and in vivo. Mechanistically, we propose a positive feedback loop mechanism. These findings provide valuable insights into the molecular pathways driving pancreatic cancer progression.
Collapse
Affiliation(s)
- Qin Xu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Shujie Dong
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiuyi Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Qun Dai
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Rubin Cheng
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqing Ge
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
49
|
Xie B, Zhong B, Zhao Z, Hu J, Yang J, Xie Y, Zhang J, Long J, Yang X, Li H. STEAP4 inhibits cisplatin-induced chemotherapy resistance through suppressing PI3K/AKT in hepatocellular carcinoma. Cancer Metab 2023; 11:26. [PMID: 38111065 PMCID: PMC10726618 DOI: 10.1186/s40170-023-00323-1] [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: 06/16/2023] [Accepted: 11/04/2023] [Indexed: 12/20/2023] Open
Abstract
Chemotherapy resistance is the leading cause for hepatocellular carcinoma (HCC)-induced death. Exploring resistance generation mechanism is an urgent need for HCC therapy. Here, we found STEAP4 was significantly downregulated in HCC patients with recurrence. Patients with low STEAP4 had poor outcome, suggesting STEAP4 might inhibit chemotherapy resistance. Cell viability assay, colony formation assay, apoptosis assay, soft agar growth assay, and tumor animal model showed STEAP4 inhibited cisplatin resistance. Mechanism analysis showed STEAP4 inhibited PI3K/AKT pathway through directly interacting with AKT. Double knockdown of STEP4 and AKT significantly inhibited cisplatin resistance. We also found STEAP4 expression was negatively correlated with PI3K/AKT pathway activity in clinic specimens. In summary, our findings suggested STEAP4 inhibited cisplatin resistance through suppressing PI3K/AKT pathway activity, providing a target for HCC therapy.
Collapse
Affiliation(s)
- Binhui Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
- Ganzhou Key Laboratory of Hepatocellular Carcinoma, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Baiyin Zhong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
- Ganzhou Key Laboratory of Hepatocellular Carcinoma, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Zhenxian Zhao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Jie Hu
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Jianqiong Yang
- Department of Clinical Research Center, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Yuankang Xie
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
- Ganzhou Key Laboratory of Hepatocellular Carcinoma, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Jianhong Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
- Ganzhou Key Laboratory of Hepatocellular Carcinoma, the First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Jianting Long
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, People's Republic of China.
| | - Xuewei Yang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, People's Republic of China.
| | - Heping Li
- Department of Medical Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
50
|
Xie Y, Zhao G, Lei X, Cui N, Wang H. Advances in the regulatory mechanisms of mTOR in necroptosis. Front Immunol 2023; 14:1297408. [PMID: 38164133 PMCID: PMC10757967 DOI: 10.3389/fimmu.2023.1297408] [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: 09/20/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
The mammalian target of rapamycin (mTOR), an evolutionarily highly conserved serine/threonine protein kinase, plays a prominent role in controlling gene expression, metabolism, and cell death. Programmed cell death (PCD) is indispensable for maintaining homeostasis by removing senescent, defective, or malignant cells. Necroptosis, a type of PCD, relies on the interplay between receptor-interacting serine-threonine kinases (RIPKs) and the membrane perforation by mixed lineage kinase domain-like protein (MLKL), which is distinguished from apoptosis. With the development of necroptosis-regulating mechanisms, the importance of mTOR in the complex network of intersecting signaling pathways that govern the process has become more evident. mTOR is directly responsible for the regulation of RIPKs. Autophagy is an indirect mechanism by which mTOR regulates the removal and interaction of RIPKs. Another necroptosis trigger is reactive oxygen species (ROS) produced by oxidative stress; mTOR regulates necroptosis by exploiting ROS. Considering the intricacy of the signal network, it is reasonable to assume that mTOR exerts a bifacial effect on necroptosis. However, additional research is necessary to elucidate the underlying mechanisms. In this review, we summarized the mechanisms underlying mTOR activation and necroptosis and highlighted the signaling pathway through which mTOR regulates necroptosis. The development of therapeutic targets for various diseases has been greatly advanced by the expanding knowledge of how mTOR regulates necroptosis.
Collapse
Affiliation(s)
- Yawen Xie
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Guoyu Zhao
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xianli Lei
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Cui
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hao Wang
- Department of Critical Care Medicine, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|