1
|
Sabeel Z, Wang J, Dong J, Liu Y, Yu C, Yang Z. The duality of GSK-3β in urinary bladder cancer: Tumor suppressor and promoter roles through multiple signaling pathways. Biochim Biophys Acta Rev Cancer 2025; 1880:189324. [PMID: 40258445 DOI: 10.1016/j.bbcan.2025.189324] [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: 11/28/2024] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/23/2025]
Abstract
Urinary bladder cancer (UBC), the tenth most common cancer globally, is primarily categorized into non-muscle-invasive (NMIBC) and muscle-invasive (MIBC) types. NMIBC has a low risk of metastasis but tends to recur frequently after transurethral resection, whereas MIBC is associated with a higher likelihood of metastasis and poorer prognosis. At diagnosis, roughly 75 % of UBC patients have NMIBC, while the remaining 25 % present with tumor invasion into the bladder's muscle layer. The molecular complexity of UBC has driven research toward identifying subtypes for more personalized treatment approaches. Glycogen synthase kinase-3β (GSK-3β) has emerged as a pivotal regulator in UBC through its dual roles across six key pathways: (1) Wnt/β-catenin regulation (tumor suppression vs oncogenic activation), (2) ER stress responses (apoptosis induction vs cytoprotection), (3) Akt/GSK-3β/β-catenin/c-Myc signaling, (4) PI3K/Akt/mTOR interactions, (5) NF-κB-mediated immune modulation, and (6) Snail1/β-catenin-driven epithelial mesenchymal transition (EMT). Our analysis reveals that GSK-3β's context-dependent functions create both therapeutic opportunities and challenges - while inhibition suppresses tumor growth via β-catenin degradation, it may simultaneously activate NF-κB-mediated oncogenic processes. These paradoxical effects are particularly evident in the tumor microenvironment, where GSK-3β modulation differentially regulates CD8+ T cell function and macrophage polarization. Understanding these complex pathway interactions is crucial for developing precision therapies that exploit GSK-3β's tumor-suppressive roles while mitigating its oncogenic potential.
Collapse
Affiliation(s)
- Zufa Sabeel
- College of Life Science and Technology, State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Jianfeng Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| | - Jian Dong
- College of Life Science and Technology, State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Yan Liu
- College of Life Science and Technology, State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Changyuan Yu
- College of Life Science and Technology, State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China.
| | - Zhao Yang
- College of Life Science and Technology, State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
2
|
Dong L, Long T, Zhang S, Mao Y, Liu M, Zhao F, Yang Z, Li L, Chen SW, Liao S, Dong Y. Structure-activity relationship explorations of 2-(isoxazol-5-yl)phenyl-3,4-dihydroxybenzoate derivatives to develop potent Wnt/β-catenin pathway inhibitors for colorectal cancer treatment. Bioorg Chem 2025; 160:108433. [PMID: 40188614 DOI: 10.1016/j.bioorg.2025.108433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/08/2025]
Abstract
In the canonical Wnt/β-catenin pathway, the nucleus translocation of β-catenin and β-catenin/ B-cell lymphoma 9 (BCL9) protein-protein interactions (PPI) promote the expressions of oncoproteins (Cyclin D1 and C-myc), thereby inducing the colorectal cancer. Herein, we report the identification of the highly potent Wnt/β-catenin pathway inhibitor 19 t following structure-activity relationship (SAR) exploration of 2-(isoxazol-5-yl)phenyl-3,4-dihydroxybenzoate which was discovered by our previous work. Further mechanism research confirmed that the optimized compound 19 t reduced the expressions of oncoproteins (Cyclin D1 and C-myc) through inhibiting the nucleus translocation of β-catenin and disrupting the interaction of β-catenin/BCL9, thereby inducing the apoptosis of SW480 cells. Encouragingly, the results of HCT116-xenograft nude mice demonstrated that the compound 19 t with acceptable pharmacokinetic parameters significantly inhibited tumor growth (TGI: 61.85 % at 20 mg/kg and 77.52 % at 40 mg/kg) and did not exhibit objective hepatotoxicity and nephrotoxicity. Consistently, the compound 19 t could also inhibit expressions of Cyclin D1 and C-myc in vivo. Collectively, the optimized compound 19 t could serve as a promising Wnt/β-catenin pathway inhibitor for colorectal cancer treatment.
Collapse
Affiliation(s)
- Li Dong
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China.
| | - Tiemei Long
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China
| | - Shanghui Zhang
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China
| | - Yongqing Mao
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China
| | - Mingji Liu
- Pharmacy Department, Guizhou Provincial People's Hospital, Nanming District, 550002, Guiyang, China
| | - Fuhui Zhao
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China
| | - Zhangxiang Yang
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China
| | - Lei Li
- Guizhou Provincial Center for Disease Control and Prevention, Guiyang 550004, China
| | - Shi-Wu Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China.
| | - Shanggao Liao
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China.
| | - Yongxi Dong
- University Engineering Research Center for the Prevention and Treatment of Chronic Diseases by Authentic Medicinal Materials in Guizhou Province & School of Pharmacy, Guizhou Medical University, Guian New District, 561113, China.
| |
Collapse
|
3
|
Zhang J, Guo H, Gong C, Shen J, Jiang G, Liu J, Liang T, Guo L. Therapeutic targets in the Wnt signaling pathway: Treating cancer with specificity. Biochem Pharmacol 2025; 236:116848. [PMID: 40049295 DOI: 10.1016/j.bcp.2025.116848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
The Wnt signaling pathway is a critical regulatory mechanism that governs cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, and the tumor immune microenvironment, while also maintaining tissue homeostasis. Dysregulated activation of this pathway is implicated in various cancers, closely linked to tumor initiation, progression, and metastasis. The Wnt/β-catenin axis plays a central role in the pathogenesis of common cancers, including colorectal cancer (CRC), breast cancer (BC), liver cancer, and lung cancer. Unlike traditional chemotherapy, targeted therapy offers a more precise approach to cancer treatment. As a key regulator of oncogenesis, the Wnt pathway represents a promising target for clinical interventions. This review provides a comprehensive analysis of the Wnt signaling pathway, exploring its roles in tumor biology and its implications in human malignancies. It further examines the molecular mechanisms and modes of action across different cancers, detailing how the Wnt pathway contributes to tumor progression through mechanisms such as metastasis promotion, immune modulation, drug resistance, and enhanced cellular proliferation. Finally, therapeutic strategies targeting Wnt pathway components are discussed, including inhibitors targeting extracellular members, as well as those within the cell membrane, cytoplasm, and nucleus. The potential of these targets in the development of novel therapeutic agents underscores the critical importance of intervening in the Wnt signaling pathway for effective cancer treatment.
Collapse
Affiliation(s)
- Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Chengxuan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jie Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guijie Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiarui Liu
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China.
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
4
|
Maurice MM, Angers S. Mechanistic insights into Wnt-β-catenin pathway activation and signal transduction. Nat Rev Mol Cell Biol 2025; 26:371-388. [PMID: 39856369 DOI: 10.1038/s41580-024-00823-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/27/2025]
Abstract
In multicellular organisms, Wnt proteins govern stem and progenitor cell renewal and differentiation to regulate embryonic development, adult tissue homeostasis and tissue regeneration. Defects in canonical Wnt signalling, which is transduced intracellularly by β-catenin, have been associated with developmental disorders, degenerative diseases and cancers. Although a simple model describing Wnt-β-catenin signalling is widely used to introduce this pathway and has largely remained unchanged over the past 30 years, in this Review we discuss recent studies that have provided important new insights into the mechanisms of Wnt production, receptor activation and intracellular signalling that advance our understanding of the molecular mechanisms that underlie this important cell-cell communication system. In addition, we review the recent development of molecules capable of activating the Wnt-β-catenin pathway with selectivity in vitro and in vivo that is enabling new lines of study to pave the way for the development of Wnt therapies for the treatment of human diseases.
Collapse
Affiliation(s)
- Madelon M Maurice
- Center for Molecular Medicine, University Medical Center, Utrecht, Netherlands.
- Oncode Institute, Utrecht, Netherlands.
| | - Stephane Angers
- Donnelly Centre for Cellular and Biomolecular Research and Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Zhang X, Li S, Hao L, Jia F, Yu F, Hu X. Influencing factors and mechanism of hepatocyte regeneration. J Transl Med 2025; 23:493. [PMID: 40307789 PMCID: PMC12042435 DOI: 10.1186/s12967-025-06278-9] [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: 10/23/2024] [Accepted: 02/20/2025] [Indexed: 05/02/2025] Open
Abstract
As a research hotspot in the field of regenerative medicine, hepatocyte regeneration has great potential in the treatment of liver diseases. This paper comprehensively summarizes the diverse sources of hepatocyte regeneration and its complex influencing factors, and deeply discusses the typical mechanism. According to the existing research, we observed that Wnt signaling pathway and Notch signaling pathway can play a synergistic role in the process of hepatocyte regeneration. So we further analyzed the crosstalk between Wnt and Notch signal pathway and the cross mechanism with TGF-β, YAP/TAZ pathway during regeneration. Despite the remarkable progress in the study of liver regeneration at the cellular and molecular levels, the comprehensive understanding of the fine regulation of influencing factors and the interaction between mechanisms still needs to be deepened. This paper aims to systematically analyze the interaction between influencing factors and classical mechanisms of hepatocyte regeneration by integrating multi-group data and advanced bioinformatics methods, so as to provide feasible ideas for the treatment of liver diseases and lay a solid theoretical foundation for the future development of regenerative medicine. It is believed that focusing on the rational development of innovative means such as inducing gene tendentiousness expression and anti-aging therapy, and in-depth analysis of the complex interactive network between hepatocyte regeneration mechanisms are expected to open up a new road for the development of more effective treatment strategies for liver diseases.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Shenghao Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Liyuan Hao
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Fukang Jia
- Henan University of Traditional Chinese, Zhengzhou, China
| | - Fei Yu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China
- Clinical Medical College of Chengdu, University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Infectious Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, No.39, Shierqiao Road, Jinniu District, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Aceves-Ewing NM, Lanza DG, Marcogliese PC, Lu D, Hsu CW, Gonzalez M, Christiansen AE, Rasmussen TL, Ho AJ, Gaspero A, Seavitt J, Dickinson ME, Yuan B, Shayota BJ, Pachter S, Hu X, Day-Salvatore DL, Mackay L, Kanca O, Wangler MF, Potocki L, Rosenfeld JA, Lewis RA, Chao HT, Lee B, Lee S, Yamamoto S, Bellen HJ, Burrage LC, Heaney JD. Uncovering Phenotypic Expansion in AXIN2-Related Disorders through Precision Animal Modeling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.05.24318524. [PMID: 39677486 PMCID: PMC11643287 DOI: 10.1101/2024.12.05.24318524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Heterozygous pathogenic variants in AXIN2 are associated with oligodontia-colorectal cancer syndrome (ODCRCS), a disorder characterized by oligodontia, colorectal cancer, and in some cases, sparse hair and eyebrows. We have identified four individuals with one of two de novo , heterozygous variants (NM_004655.4:c.196G>A, p.(Glu66Lys) and c.199G>A, p.(Gly67Arg)) in AXIN2 whose presentations expand the phenotype of AXIN2-related disorders. In addition to ODCRCS features, these individuals have global developmental delay, microcephaly, and limb, ophthalmologic, and renal abnormalities. Structural modeling of these variants suggests that they disrupt AXIN2 binding to tankyrase, which regulates AXIN2 levels through PARsylation and subsequent proteasomal degradation. To test whether these variants produce a phenotype in vivo , we utilized an innovative prime editing N1 screen to phenotype heterozygous (p.E66K) mouse embryos, which were perinatal lethal with short palate and skeletal abnormalities, contrary to published viable Axin2 null mouse models. Modeling of the p.E66K variant in the Drosophila wing revealed gain-of-function activity compared to reference AXIN2. However, the variant showed loss-of-function activity in the fly eye compared to reference AXIN2, suggesting that the mechanism by which p.E66K affects AXIN2 function is cell context-dependent. Together, our studies in humans, mice, and flies demonstrate that specific variants in the tankyrase-binding domain of AXIN2 are pathogenic, leading to phenotypic expansion with context-dependent effects on AXIN2 function and WNT signaling. Moreover, the modeling strategies used to demonstrate variant pathogenicity may be beneficial for the resolution of other de novo heterozygous variants of uncertain significance associated with congenital anomalies in humans.
Collapse
|
7
|
Karati D, Meur S, Roy S, Mukherjee S, Debnath B, Jha SK, Sarkar BK, Naskar S, Ghosh P. Glycogen synthase kinase 3 (GSK3) inhibition: a potential therapeutic strategy for Alzheimer's disease. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2319-2342. [PMID: 39432068 DOI: 10.1007/s00210-024-03500-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease (AD), the most common type of dementia among older adults, is a chronic neurodegenerative pathology that causes a progressive loss of cognitive functioning with a decline of rational skills. It is well known that AD is multifactorial, so there are many different pharmacological targets that can be pursued. According to estimates from the World Health Organization (WHO), 18 million individuals worldwide suffer from AD. Major initiatives to identify risk factors, enhance care giving, and conduct basic research to delay the beginning of AD were started by the USA, France, Germany, France, and various other nations. Widely recognized as a key player in the development and subsequent progression of AD pathogenesis, glycogen synthase kinase-3 (GSK-3) controls a number of crucial targets associated with neuronal degeneration. GSK-3 inhibition has been linked to reduced tau hyperphosphorylation, β-amyloid formation, and neuroprotective benefits in Alzheimer's disease. Lithium, the very first inhibitor of GSK-3β that was used therapeutically, has been successfully used for many years with remarkable results. A great variety of structurally varied strong GSK-3β blockers have been identified in recent years. The purpose of this thorough review is to cover the biological and structural elements of glycogen synthase kinase, as well as the medicinal chemistry aspects of GSK inhibitors that have been produced in recent years.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, West Bengal, 700091, India
| | - Shreyasi Meur
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, West Bengal, 700091, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India.
| | - Biplab Debnath
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal, 711316, India
| | - Sajal Kumar Jha
- Department of Pharmaceutical Technology, Bengal College of Pharmaceutical Technology, Dubrajpur, West Bengal, 731123, India
| | | | - Saheli Naskar
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| | - Priya Ghosh
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata - Group of Institutions, 124, B.L Saha Road, Kolkata, West Bengal, 700053, India
| |
Collapse
|
8
|
Kim J, Jeon YJ, Chang IY, Lee JH, You HJ. Disruption of the β-catenin destruction complex via Ephexin1-Axin1 interaction promotes colorectal cancer proliferation. Exp Mol Med 2025; 57:151-166. [PMID: 39741188 PMCID: PMC11799323 DOI: 10.1038/s12276-024-01381-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 01/02/2025] Open
Abstract
Wnt signaling is essential for cell growth and tumor formation and is abnormally activated in colorectal cancer (CRC), contributing to tumor progression; however, the specific role and regulatory mechanisms involved in tumor development remain unclear. Here, we show that Ephexin1, a guanine nucleotide exchange factor, is significantly overexpressed in CRC and is correlated with increased Wnt/β-catenin pathway activity. Through comprehensive analysis, including RNA sequencing data from TCGA and functional assays, we observed that Ephexin1 promotes tumor proliferation and migration by activating the Wnt/β-catenin pathway. This effect was mediated by the interaction of Ephexin1 with Axin1, a critical component of the β-catenin destruction complex, which in turn enhanced the stability and activity of β-catenin in signaling pathways critical for tumor development. Importantly, our findings also suggest that targeting Ephexin1 may increase the efficacy of Wnt/β-catenin pathway inhibitors in CRC treatment. These findings highlight the potential of targeting Ephexin1 as a strategy for developing effective treatments for CRC, suggesting a novel and promising approach to therapy aimed at inhibiting cancer progression.
Collapse
Affiliation(s)
- Jeeho Kim
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea
- Department of Pharmacology, Gwangju, South Korea
| | | | | | - Jung-Hee Lee
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Cellular and Molecular Medicine, Chosun University School of Medicine, 375 Seosuk-dong, Gwangju, 501-759, South Korea.
| | - Ho Jin You
- Laboratory of Genomic Instability and Cancer Therapeutics, Gwangju, South Korea.
- Department of Pharmacology, Gwangju, South Korea.
| |
Collapse
|
9
|
Onji M, Sigl V, Lendl T, Novatchkova M, Ullate-Agote A, Andersson-Rolf A, Kozieradzki I, Koglgruber R, Pai TP, Lichtscheidl D, Nayak K, Zilbauer M, Carranza García NA, Sievers LK, Falk-Paulsen M, Cronin SJF, Hagelkruys A, Sawa S, Osborne LC, Rosenstiel P, Pasparakis M, Ruland J, Takayanagi H, Clevers H, Koo BK, Penninger JM. RANK drives structured intestinal epithelial expansion during pregnancy. Nature 2025; 637:156-166. [PMID: 39633049 PMCID: PMC11666467 DOI: 10.1038/s41586-024-08284-1] [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: 03/10/2023] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
During reproduction, multiple species such as insects and all mammals undergo extensive physiological and morphological adaptions to ensure health and survival of the mother and optimal development of the offspring. Here we report that the intestinal epithelium undergoes expansion during pregnancy and lactation in mammals. This enlargement of the intestinal surface area results in a novel geometry of expanded villi. Receptor activator of nuclear factor-κΒ (RANK, encoded by TNFRSF11A) and its ligand RANKL were identified as a molecular pathway involved in this villous expansion of the small intestine in vivo in mice and in intestinal mouse and human organoids. Mechanistically, RANK-RANKL protects gut epithelial cells from cell death and controls the intestinal stem cell niche through BMP receptor signalling, resulting in the elongation of villi and a prominent increase in the intestinal surface. As a transgenerational consequence, babies born to female mice that lack Rank in the intestinal epithelium show reduced weight and develop glucose intolerance after metabolic stress. Whereas gut epithelial remodelling in pregnancy/lactation is reversible, constitutive expression of an active form of RANK is sufficient to drive intestinal expansion followed by loss of villi and stem cells, and prevents the formation of Apcmin-driven small intestinal stem cell tumours. These data identify RANK-RANKL as a pathway that drives intestinal epithelial expansion in pregnancy/lactation, one of the most elusive and fundamental tissue remodelling events in mammalian life history and evolution.
Collapse
Affiliation(s)
- Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| | - Verena Sigl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Thomas Lendl
- Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Asier Ullate-Agote
- Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amanda Andersson-Rolf
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, The Netherlands
| | - Ivona Kozieradzki
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rubina Koglgruber
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Tsung-Pin Pai
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Dominic Lichtscheidl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Komal Nayak
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Matthias Zilbauer
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke's, Cambridge, UK
| | - Natalia A Carranza García
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Katharina Sievers
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Astrid Hagelkruys
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Shinichiro Sawa
- Division of Mucosal Immunology, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Lisa C Osborne
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Manolis Pasparakis
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, TUM University Hospital, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, The Netherlands
- The Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, Basel, Switzerland
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Republic of Korea
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
10
|
Han Y, Gao H, Gao J, Yang Y, He C. Low-intensity pulsed ultrasound regulates bone marrow mesenchymal stromal cells differentiation and inhibits bone loss by activating the IL-11-Wnt/β-catenin signaling pathway. Int Immunopharmacol 2024; 143:113380. [PMID: 39405933 DOI: 10.1016/j.intimp.2024.113380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a common metabolic bone disease. Low-intensity pulsed ultrasound (LIPUS) can effectively promote bone formation and fracture healing. The Wnt/β-catenin signaling pathway is crucial for regulating bone homeostasis and bone diseases, and its downregulation is one of the main mechanisms of osteoporosis pathogenesis. Interleukin-11 (IL-11), which is regulated by mechanical stress, is a key factor in bone remodeling. Here, we investigated the optimal intervention parameters for LIPUS, the relationships among LIPUS, IL-11, and the Wnt/β-catenin signaling pathway, and the effects of LIPUS on bone loss and potential molecular mechanisms in ovariectomized (OVX) mice. METHODS Bone marrow mesenchymal stromal cells (BMSCs) were subjected to LIPUS intervention for 0, 10, or 20 min to determine the optimal intervention time. The mediating role of IL-11 in LIPUS intervention was explored through IL-11 knockdown and overexpression. Finally, animal experiments were conducted to investigate the in vivo therapeutic effects of LIPUS. RESULTS The optimal intervention time for LIPUS was 20 min. LIPUS promoted IL-11 expression and upregulated the Wnt/β-catenin signaling pathway, thereby promoting osteogenic differentiation and inhibiting adipogenic differentiation of BMSCs. IL-11 mediates the regulation of the Wnt/β-catenin signaling pathway by LIPUS. Additionally, LIPUS effectively improved the bone microstructure in ovariectomized mice, inhibited bone loss, promoted IL-11 expression in bone tissue, and activated the Wnt/β-catenin signaling pathway in the femur. CONCLUSION Low-intensity pulsed ultrasound can regulate BMSCs differentiation and inhibit bone loss by promoting IL-11 expression and activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yijing Han
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Hui Gao
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000
| | - Jing Gao
- LIFU Medical Research Center, Sichuan Taiyou Technology Co., Ltd., Chengdu, Sichuan, China, 610000
| | - Yonghong Yang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China, 610000; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China, 610000.
| |
Collapse
|
11
|
Saadh MJ, Khalifehsoltani A, Hussein AHA, Allela OQB, Sameer HN, Rizaev J, Hameed HG, Idan AH, Alsaikhan F. Exosomal microRNAs in cancer metastasis: A bridge between tumor micro and macroenvironment. Pathol Res Pract 2024; 263:155666. [PMID: 39476605 DOI: 10.1016/j.prp.2024.155666] [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: 09/25/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 11/10/2024]
Abstract
Malignant tumors are complicated structures of cancer cells that are constantly in communication with their local and distant environment. Exosomes are released by tumor cells and can facilitate the cell-cell interaction within the local microenvironment and the primary tumor. In fact, exosomes are secreted by both tumor and non-tumor cells, to provide a mutual communication network between cells and their micro- and/or macro-environments. Exososmes can contain a variety of biological cargos mostly based on their originated cells. Uptake of these exosomes by their recipient cells results in the alterations that their cargo can exert. MicroRNAs are identified as one of the most critical exosomal components, considering their pivotal regulatory roles in distinct biological process, including metastasis. Release and absorbance of exosomal microRNAs is possible by various cells within the host, and can have distinct biological consequences. Therefore, in this review we will discuss the role of exosomal microRNAs derived from tumor cells and untransformed cells within their micro- and macroenvironment in cancer progression and metastasis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan
| | | | | | | | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq
| | - Jasur Rizaev
- Department of Public health and Healthcare management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | | | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia; School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| |
Collapse
|
12
|
Rodvold JJ, Grimmer M, Ruiz K, Marsters SA, Oikonomidi I, Tan-Aristy E, Pham VC, Sarkar T, Harnoss JM, Shatz-Binder W, Modrusan ZD, Wu TD, Lill JR, Villemure E, Rudolph J, de Sousa e Melo F, Ashkenazi A. ATF6 Promotes Colorectal Cancer Growth and Stemness by Regulating the Wnt Pathway. CANCER RESEARCH COMMUNICATIONS 2024; 4:2734-2755. [PMID: 39324706 PMCID: PMC11492184 DOI: 10.1158/2767-9764.crc-24-0268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/07/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
SIGNIFICANCE ATF6 intervention reduces colorectal cancer cell and organoid viability by interrupting dysregulated Wnt signaling, identifying a novel facilitator and potential therapeutic target in colorectal cancer.
Collapse
Affiliation(s)
- Jeffrey J. Rodvold
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Matthew Grimmer
- Department of Computational Science, Genentech, Inc., South San Francisco, California
| | - Karen Ruiz
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California
| | - Scot A. Marsters
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Ioanna Oikonomidi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Eileen Tan-Aristy
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| | - Victoria C. Pham
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Tamal Sarkar
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University Hospital Giessen, Giessen, Germany
| | - Jonathan M. Harnoss
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University Hospital Giessen, Giessen, Germany
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., South San Francisco, California
| | - Zora D. Modrusan
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Thomas D. Wu
- Department of Computational Science, Genentech, Inc., South San Francisco, California
| | - Jennie R. Lill
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, California
| | - Elisia Villemure
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | - Joachim Rudolph
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California
| | | | - Avi Ashkenazi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California
| |
Collapse
|
13
|
Sun S, Li H, Liu S, Xie X, Zhai W, Pan J. Long noncoding RNA UCA1 inhibits epirubicin-induced apoptosis by activating PPARα-mediated lipid metabolism. Exp Cell Res 2024; 442:114271. [PMID: 39357639 DOI: 10.1016/j.yexcr.2024.114271] [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: 08/15/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Metabolic reprogramming is a hallmark of cancer, and abnormal lipid metabolism is associated with drug resistance in bladder cancer cells. The long noncoding RNA (lncRNA) UCA1 is overexpressed in bladder cancer, but its functional contribution to lipid metabolism remains uncharacterized. In this study, we demonstrated that lncRNA UCA1 inhibits epirubicin-induced cell apoptosis by supporting abnormal lipid metabolism in bladder cancer cells. Mechanistically, lncRNA UCA1 promotes lipid accumulation in vitro and in vivo by upregulating PPARα mRNA and protein expression, which is mediated by miR-30a-3p. Knockdown of lncRNA UCA1 increased epirubicin-induced apoptosis via miR-30a-3p/PPARα and downstream p-AKT/p-GSK-3β/β-catenin signaling. Furthermore, mixed free fatty acids upregulated lncRNA UCA1 expression by promoting recruitment of the transcription factor RXRα to the lncRNA UCA1 promoter. These findings were verified in a mouse xenograft model and are consistent with the expression patterns in human bladder cancer patients. Overall, these findings establish the role of lncRNA UCA1 in lipid metabolism and bladder cancer cell resistance to epirubicin, suggesting that lncRNA UCA1 may serve as a candidate target for enhancing bladder cancer chemotherapy.
Collapse
Affiliation(s)
- Shuaijie Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University and the Key Clinical Laboratory of Henan Province, Zhengzhou, China
| | - Huijin Li
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Shanshan Liu
- Department of Clinical Laboratory, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaojuan Xie
- Shaanxi Center for Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Wen Zhai
- Department of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Jingjing Pan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University and the Key Clinical Laboratory of Henan Province, Zhengzhou, China.
| |
Collapse
|
14
|
Masci D, Puxeddu M, Silvestri R, La Regina G. Targeting CBP and p300: Emerging Anticancer Agents. Molecules 2024; 29:4524. [PMID: 39407454 PMCID: PMC11482477 DOI: 10.3390/molecules29194524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
CBP and p300 are versatile transcriptional co-activators that play essential roles in regulating a wide range of signaling pathways, including Wnt/β-catenin, p53, and HIF-1α. These co-activators influence various cellular processes such as proliferation, differentiation, apoptosis, and response to hypoxia, making them pivotal in normal physiology and disease progression. The Wnt/β-catenin signaling pathway, in particular, is crucial for cellular proliferation, differentiation, tissue homeostasis, and embryogenesis. Aberrant activation of this pathway is often associated with several types of cancer, such as colorectal tumor, prostate cancer, pancreatic and hepatocellular carcinomas. In recent years, significant efforts have been directed toward identifying and developing small molecules as novel anticancer agents capable of specifically inhibiting the interaction between β-catenin and the transcriptional co-activators CBP and p300, which are required for Wnt target gene expression and are consequently involved in the regulation of tumor cell proliferation, migration, and invasion. This review summarizes the most significant and original research articles published from 2010 to date, found by means of a PubMed search, highlighting recent advancements in developing both specific and non-specific inhibitors of CBP/β-catenin and p300/β-catenin interactions. For a more comprehensive view, we have also explored the therapeutic potential of CBP/p300 bromodomain and histone acetyltransferase inhibitors in disrupting the transcriptional activation of genes involved in various signaling pathways related to cancer progression. By focusing on these therapeutic strategies, this review aims to offer a detailed overview of recent approaches in cancer treatment that selectively target CBP and p300, with particular emphasis on their roles in Wnt/β-catenin-driven oncogenesis.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (R.S.)
| |
Collapse
|
15
|
Kalairaj MS, Pradhan R, Saleem W, Smith MM, Gaharwar AK. Intra-Articular Injectable Biomaterials for Cartilage Repair and Regeneration. Adv Healthc Mater 2024; 13:e2303794. [PMID: 38324655 PMCID: PMC11468459 DOI: 10.1002/adhm.202303794] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
Osteoarthritis is a degenerative joint disease characterized by cartilage deterioration and subsequent inflammatory changes in the underlying bone. Injectable hydrogels have emerged as a promising approach for controlled drug delivery in cartilage therapies. This review focuses on the latest developments in utilizing injectable hydrogels as vehicles for targeted drug delivery to promote cartilage repair and regeneration. The pathogenesis of osteoarthritis is discussed to provide a comprehensive understanding of the disease progression. Subsequently, the various types of injectable hydrogels used for intra-articular delivery are discussed. Specifically, physically and chemically crosslinked injectable hydrogels are critically analyzed, with an emphasis on their fabrication strategies and their capacity to encapsulate and release therapeutic agents in a controlled manner. Furthermore, the potential of incorporating growth factors, anti-inflammatory drugs, and cells within these injectable hydrogels are discussed. Overall, this review offers a comprehensive guide to navigating the landscape of hydrogel-based therapeutics in osteoarthritis.
Collapse
Affiliation(s)
| | - Ridhi Pradhan
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Waqas Saleem
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Morgan M. Smith
- Department of Veterinary Integrative BiosciencesSchool of Veterinary Medicine and Biomedical SciencesTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Genetics and Genomics Interdisciplinary ProgramTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
16
|
Gelmi MC, de Ru AH, van Veelen PA, Tjokrodirijo RTN, Stern MH, Houy A, Verdijk RM, Vu THK, Ksander BR, Vaarwater J, Kilic E, Brosens E, Jager MJ. Protein and mRNA Expression in Uveal Melanoma Cell Lines Are Related to GNA and BAP1 Mutation Status. Invest Ophthalmol Vis Sci 2024; 65:37. [PMID: 39042403 PMCID: PMC11268447 DOI: 10.1167/iovs.65.8.37] [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: 04/26/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Purpose Cell lines are being used in preclinical uveal melanoma (UM) research. Because not all cell lines harbor typical GNAQ or GNA11 hotspot mutations, we aimed at better classifying them and determining whether we could find genetic causes to explain the protein and mRNA expression profiles of the cell lines. Methods We studied protein and mRNA expression of 14 UM cell lines and determined the presence of single nucleotide variants and small insertions and deletions with next-generation sequencing and copy number alterations with a single nucleotide polymorphism array. The lists of differentially expressed proteins and genes were merged, and shared lists were created, keeping only terms with concordant mRNA and protein expression. Enrichment analyses were performed on the shared lists. Results Cell lines Mel285 and Mel290 are separate from GNA-mutated cell lines and show downregulation of melanosome-related markers. Both lack typical UM mutations but each harbors four putatively deleterious variants in CTNNB1, PPP1R10, LIMCH1, and APC in Mel285 and ARID1A, PPP1R10, SPG11, and RNF43 in Mel290. The upregulated terms in Mel285 and Mel290 did not point to a convincing alternative origin. Mel285 shows loss of chromosomes 1p, 3p, partial 3q, 6, and partial 8p, whereas Mel290 shows loss of 1p and 6. Expression in the other 12 cell lines was related to BAP1 expression. Conclusions Although Mel285 and Mel290 have copy number alterations that fit UM, multi-omics analyses show that they belong to a separate group compared to the other analyzed UM cell lines. Therefore, they may not be representative models to test potential therapeutic targets for UM.
Collapse
Affiliation(s)
- Maria Chiara Gelmi
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Arnoud H. de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A. van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marc-Henri Stern
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Alexandre Houy
- Inserm U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Institut Curie, PSL Research University, Paris, France
| | - Robert M. Verdijk
- Department of Pathology, Ophthalmic Pathology Section, Erasmus MC, Rotterdam, The Netherlands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - T. H. Khanh Vu
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruce R. Ksander
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States
| | - Jolanda Vaarwater
- Department of Clinical Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
| | - Emine Kilic
- Department of Ophthalmology, Erasmus MC, Rotterdam, The Netherlands
| | - Erwin Brosens
- Department of Clinical Genetics, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
17
|
Gholam Azad M, Hussaini M, Russell TM, Richardson V, Kaya B, Dharmasivam M, Richardson DR. Multi-modal mechanisms of the metastasis suppressor, NDRG1: Inhibition of WNT/β-catenin signaling by stabilization of protein kinase Cα. J Biol Chem 2024; 300:107417. [PMID: 38815861 PMCID: PMC11261793 DOI: 10.1016/j.jbc.2024.107417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
The metastasis suppressor, N-myc downstream regulated gene-1 (NDRG1), inhibits pro-oncogenic signaling in pancreatic cancer (PC). This investigation dissected a novel mechanism induced by NDRG1 on WNT/β-catenin signaling in multiple PC cell types. NDRG1 overexpression decreased β-catenin and downregulated glycogen synthase kinase-3β (GSK-3β) protein levels and its activation. However, β-catenin phosphorylation at Ser33, Ser37, and Thr41 are classically induced by GSK-3β was significantly increased after NDRG1 overexpression, suggesting a GSK-3β-independent mechanism. Intriguingly, NDRG1 overexpression upregulated protein kinase Cα (PKCα), with PKCα silencing preventing β-catenin phosphorylation at Ser33, Ser37, and Thr41, and decreasing β-catenin expression. Further, NDRG1 and PKCα were demonstrated to associate, with PKCα stabilization occurring after NDRG1 overexpression. PKCα half-life increased from 1.5 ± 0.8 h (3) in control cells to 11.0 ± 2.5 h (3) after NDRG1 overexpression. Thus, NDRG1 overexpression leads to the association of NDRG1 with PKCα and PKCα stabilization, resulting in β-catenin phosphorylation at Ser33, Ser37, and Thr41. The association between PKCα, NDRG1, and β-catenin was identified, with the formation of a potential metabolon that promotes the latter β-catenin phosphorylation. This anti-oncogenic activity of NDRG1 was multi-modal, with the above mechanism accompanied by the downregulation of the nucleo-cytoplasmic shuttling protein, p21-activated kinase 4 (PAK4), which is involved in β-catenin nuclear translocation, inhibition of AKT phosphorylation (Ser473), and decreased β-catenin phosphorylation at Ser552 that suppresses its transcriptional activity. These mechanisms of NDRG1 activity are important to dissect to understand the marked anti-cancer efficacy of NDRG1-inducing thiosemicarbazones that upregulate PKCα and inhibit WNT signaling.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mohammed Hussaini
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
18
|
Kuntze A, Meliß RR, Ermert L, Falkenberg KD, Puller AC, Trautmann M, Hartmann W, Wardelmann E. Superficial fibromas with CTNNB1 mutation. Genes Chromosomes Cancer 2024; 63:e23247. [PMID: 38757718 DOI: 10.1002/gcc.23247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024] Open
Abstract
Superficial fibromas are a group of mesenchymal spindle cell lesions with pathomorphological heterogeneity and diverse molecular backgrounds. In part, they may be indicators of an underlying syndrome. Among the best-known entities of superficial fibromas is Gardner fibroma, a plaque-like benign tumor, which is associated with APC germline mutations and occurs in patients with familial adenomatosis polyposis (Gardner syndrome). Affected patients also have an increased risk to develop desmoid fibromatosis (DTF), a locally aggressive neoplasm of the deep soft tissue highly prone to local recurrences. Although a minority of DTFs occur in the syndromic context and harbor APC germline mutations, most frequently their underlying molecular aberration is a sporadic mutation in Exon 3 of the CTNNB1 gene. Up to date, a non-syndromic equivalent to Gardner fibroma carrying a CTNNB1 mutation has not been defined. Here, we present two cases of (sub-)cutaneous tumors with a hypocellular and collagen-rich Gardner fibroma-like appearance and pathogenic, somatic CTNNB1 mutations. We aim to differentiate these tumors from other fibromas according to their histological appearance, immunohistochemical staining profile and underlying somatic CTNNB1 mutations. Furthermore, we distinguish them from locally aggressive desmoid fibromatosis regarding their biological behavior, prognosis and indicated therapeutic strategies. Consequently, we call them CTNNB1-mutated superficial fibromas as a sporadic counterpart lesion to syndromic Gardner fibromas.
Collapse
Affiliation(s)
- Anna Kuntze
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - R R Meliß
- Institute of Pathology and Dermatopathology, Hannover, Germany
| | - L Ermert
- Institute of Pathology, Oldenburg, Germany
| | - K D Falkenberg
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - A C Puller
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - M Trautmann
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - W Hartmann
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - E Wardelmann
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| |
Collapse
|
19
|
Aoki K, Nitta A, Igarashi A. NELF and PAF1C complexes are core transcriptional machineries controlling colon cancer stemness. Oncogene 2024; 43:566-577. [PMID: 38182897 PMCID: PMC10873196 DOI: 10.1038/s41388-023-02930-0] [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: 07/15/2023] [Revised: 12/01/2023] [Accepted: 12/20/2023] [Indexed: 01/07/2024]
Abstract
Mutations in APC, found in 80% of colon caner, enhance β-catenin stabilization, which is the initial step of colonic tumorigenesis. However, the core transcriptional mechanism underlying the induction of colon cancer stemness by stable β-catenin remains unclear. Here, we found that inducible inhibition of β-catenin suppressed elongation of Pol II and RNA polymerase-associated factor 1 complex (PAF1C) around the transcription start site (TSS) of LGR5. Moreover, stable β-catenin enhanced the formation of active Pol II complex cooperatively with CDC73 and CDK9 by facilitating the recruitment of DRB sensitivity-inducing factor (DSIF) and negative elongation factor (NELF) complexes to the Pol II complex. Subsequently, stable β-catenin facilitated the formation of the Pol II-DSIF-PAF1C complex, suggesting that stable β-catenin induces cancer stemness by stimulating active Pol II complex through NELF and PAF1C. Furthermore, NELF or PAF1C inhibition recapitulated the changes in cancer stemness-related gene expression induced by the inhibition of stable β-catenin and suppressed colon cancer stemness. Additionally, the chemical inhibition of CDK12 (a downstream transcription CDK of PAF1C) suppressed colon cancer stemness. These results suggest that NELF and PAF1C are the core transcriptional machineries that control expression of colon cancer stemness-inducing genes and may be therapeutic targets for colon cancer.
Collapse
Affiliation(s)
- Koji Aoki
- Department of Pharmacology, Faculty of Medicine, University of Fukui, Fukui, Japan.
| | - Akari Nitta
- Department of Pharmacology, Faculty of Medicine, University of Fukui, Fukui, Japan
| | - Ayumi Igarashi
- Department of Pharmacology, Faculty of Medicine, University of Fukui, Fukui, Japan
| |
Collapse
|
20
|
Yang J, Liu J, Liang J, Li F, Wang W, Chen H, Xie X. Epithelial-mesenchymal transition in age-associated thymic involution: Mechanisms and therapeutic implications. Ageing Res Rev 2023; 92:102115. [PMID: 37922996 DOI: 10.1016/j.arr.2023.102115] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/17/2023] [Accepted: 10/29/2023] [Indexed: 11/07/2023]
Abstract
The thymus is a critical immune organ with endocrine and immune functions that plays important roles in the physiological and pathological processes of the body. However, with aging, the thymus undergoes degenerative changes leading to decreased production and output of naive T cells and the secretion of thymic hormones and related cytokines, thereby promoting the occurrence and development of various age-associated diseases. Therefore, identifying essential processes that regulate age-associated thymic involution is crucial for long-term control of thymic involution and age-associated disease progression. Epithelial-mesenchymal transition (EMT) is a well-established process involved in organ aging and functional impairment through tissue fibrosis in several organs, such as the heart and kidney. In the thymus, EMT promotes fibrosis and potentially adipogenesis, leading to thymic involution. This review focuses on the factors involved in thymic involution, including oxidative stress, inflammation, and hormones, from the perspective of EMT. Furthermore, current interventions for reversing age-associated thymic involution by targeting EMT-associated processes are summarized. Understanding the key mechanisms of thymic involution through EMT as an entry point may promote the development of new therapies and clinical agents to reverse thymic involution and age-associated disease.
Collapse
Affiliation(s)
- Jiali Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Juan Liu
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Jiayu Liang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Fan Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Wenwen Wang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China; Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China.
| |
Collapse
|
21
|
Farnhammer F, Colozza G, Kim J. RNF43 and ZNRF3 in Wnt Signaling - A Master Regulator at the Membrane. Int J Stem Cells 2023; 16:376-384. [PMID: 37643759 PMCID: PMC10686798 DOI: 10.15283/ijsc23070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/10/2023] [Indexed: 08/31/2023] Open
Abstract
The Wnt β-catenin signaling pathway is a highly conserved mechanism that plays a critical role from embryonic development and adult stem cell homeostasis. However, dysregulation of the Wnt pathway has been implicated in various diseases, including cancer. Therefore, multiple layers of regulatory mechanisms tightly control the activation and suppression of the Wnt signal. The E3 ubiquitin ligases RNF43 and ZNRF3, which are known negative regulators of the Wnt pathway, are critical component of Wnt signaling regulation. These E3 ubiquitin ligases control Wnt signaling by targeting the Wnt receptor Frizzled to induce ubiquitination-mediated endo-lysosomal degradation, thus controlling the activation of the Wnt signaling pathway. We also discuss the regulatory mechanisms, interactors, and evolution of RNF43 and ZNRF3. This review article summarizes recent findings on RNF43 and ZNRF3 and their potential implications for the development of therapeutic strategies to target the Wnt signaling pathway in various diseases, including cancer.
Collapse
Affiliation(s)
- Fiona Farnhammer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
- Division of Oncology and Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Jihoon Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Korea
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Korea
| |
Collapse
|
22
|
Odintsov I, Dong F, Guenette JP, Fritchie KJ, Jo VY, Fletcher CDM, Papke DJ. Infantile Sinonasal Myxoma Is Clinically and Genetically Distinct From Other Myxomas of the Craniofacial Bones and From Desmoid Fibromatosis. Am J Surg Pathol 2023; 47:1301-1315. [PMID: 37678343 DOI: 10.1097/pas.0000000000002119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Sinonasal myxomas are rare benign tumors of the maxillary bone and sinus. There is published evidence that sinonasal myxomas occurring in children up to 3 years of age ("infantile sinonasal myxomas") are clinically distinctive and harbor Wnt signaling pathway alterations. Here, we characterized 16 infantile sinonasal myxomas and compared them to 19 maxillary myxomas and 11 mandibular myxomas in older patients. Clinical follow-up was available for 21 patients (46%) overall (median: 2.6 y; range: 4 mo to 21 y), including 10 of 16 infantile sinonasal myxomas (62%). None of the 8 resected infantile sinonasal myxomas recurred, despite positive margins in 6 of them. One incompletely resected infantile sinonasal myxoma underwent partial regression without additional treatment. In contrast, 4 of the 11 other myxomas with follow-up recurred (36%), including one that recurred twice. Imaging studies demonstrated all infantile sinonasal myxomas to be expansile lesions arising from the anterior maxillary bone adjacent to the nasal aperture, with peripheral reactive bone formation. Histologically, infantile sinonasal myxomas showed short, intersecting fascicles of bland fibroblastic cells with prominent stromal vessels. Examples with collagenous stroma showed some morphologic overlap with desmoid fibromatosis, although none showed infiltrative growth into adjacent soft tissue. Immunohistochemistry demonstrated nuclear β-catenin expression in 14 of 15 infantile sinonasal myxomas (93%), in contrast to 4 of 26 other myxomas of craniofacial bones (15%). Smooth muscle actin was expressed in only 1 of 11 infantile sinonasal myxomas (9%). Next-generation sequencing was successfully performed on 10 infantile sinonasal myxomas and 7 other myxomas. Infantile sinonasal myxomas harbored CTNNB1 point mutations in 4 cases (D32Y, G34E, G34R, and I35S), and none harbored alterations to the phosphorylation sites T41 and S45 that are altered in 99% of CTNNB1 -mutant desmoid fibromatoses. Three tumors showed alterations consistent with biallelic APC inactivation. Three infantile sinonasal myxomas that showed strong nuclear β-catenin expression were negative for CTNNB1 and APC alterations. Sequencing was negative for CTNNB1 or APC alterations in all 7 myxomas of craniofacial bones in older patients. Four of these myxomas in older patients (57%) showed copy number alterations, and all lacked known driving alterations. These findings support the notion that infantile sinonasal myxomas are clinically and genetically distinctive, and we propose the use of the diagnostic term "infantile sinonasal myxoma" to distinguish this tumor type from other myxomas of the craniofacial bones. Infantile sinonasal myxoma should be distinguished from desmoid fibromatosis because of its unique clinical presentation, more indolent clinical behavior, different morphology, different immunohistochemical profile, and different genetics. Given its indolent behavior even when marginally excised, infantile sinonasal myxoma can be managed with conservative surgery.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| | - Jeffrey P Guenette
- Division of Neuroradiology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA
| | | | - Vickie Y Jo
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| | | | - David J Papke
- Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston
| |
Collapse
|
23
|
Che Y, Zhang H, Li H, Wu X. CIP2A interacts with AKT1 to promote the malignant biological behaviors of oral squamous cell carcinoma by upregulating the GSK‑3β/β‑catenin pathway. Exp Ther Med 2023; 26:514. [PMID: 37840566 PMCID: PMC10570767 DOI: 10.3892/etm.2023.12213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/18/2023] [Indexed: 10/17/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common malignancies worldwide, which is associated with a poor prognosis. The present study aimed to investigate the role of cancerous inhibitor of protein phosphatase 2A (CIP2A) in OSCC and its regulatory effect on AKT1. Firstly, CIP2A and AKT1 expression in OSCC cells was detected by western blotting. After silencing CIP2A, cell viability and cell proliferation were assessed using the Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine staining. Cell apoptosis was evaluated by TUNEL staining and the expression of apoptosis-related proteins was assessed using western blotting. Wound healing, Transwell and tube formation assays were performed to evaluate CAL-27 cell migration, invasion and human umbilical vein endothelial cell (HUVEC) tube formation. The interaction between CIP2A and AKT1 was identified by co-immunoprecipitation (co-IP). In addition, AKT1 was overexpressed in CIP2A-silenced CAL-27 cells to perform rescue experiments to analyze the malignant biological functions of CAL-27 cells. Finally, the expression of proteins in the glycogen synthase kinase (GSK)-3β/β-catenin pathway was determined by western blot analysis. Markedly elevated CIP2A and AKT1 expression was observed in OSCC cells. CIP2A knockdown inhibited the viability, proliferation, migration and invasion, and promoted the apoptosis of CAL-27 cells. Concurrently, CIP2A loss-of-function attenuated tube formation. Results of Co-IP confirmed there was an interaction between CIP2A and AKT1. Rescue experiments suggested that AKT1 overexpression alleviated the inhibitory effects of CIP2A knockdown on the viability, proliferation, migration and invasion of CAL-27 cells, as well as tube formation in HUVECs . Additionally, CIP2A silencing significantly downregulated phosphorylated-GSK-3β and β-catenin expression, which was reversed by AKT1 overexpression. In conclusion, CIP2A could interact with AKT1 to promote the malignant biological behaviors of OSCC cells by upregulating the GSK-3β/β-catenin pathway. These findings may provide a targeted therapy for OSCC treatment.
Collapse
Affiliation(s)
- Yilei Che
- Department of Stomatology, Aerospace Center Hospital, Beijing 100049, P.R. China
| | - Hui Zhang
- Department of Stomatology, Aerospace Center Hospital, Beijing 100049, P.R. China
| | - Hui Li
- Department of Stomatology, Aerospace Center Hospital, Beijing 100049, P.R. China
| | - Xiaozhen Wu
- Department of Stomatology, Aerospace Center Hospital, Beijing 100049, P.R. China
| |
Collapse
|
24
|
Tse J, O’Keefe R, Rigopolous A, Carli ALE, Waaler J, Krauss S, Ernst M, Buchert M. A Mouse Model for the Rapid and Binomial Assessment of Putative WNT/β-Catenin Signalling Inhibitors. Biomedicines 2023; 11:2719. [PMID: 37893093 PMCID: PMC10604108 DOI: 10.3390/biomedicines11102719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Specific signalling thresholds of the WNT/β-catenin pathway affect embryogenesis and tissue homeostasis in the adult, with mutations in this pathway frequently occurring in cancer. Excessive WNT/β-catenin activity inhibits murine anterior development associated with embryonic lethality and accounts for the driver event in 80% of human colorectal cancers. Uncontrolled WNT/β-catenin signalling arises primarily from impairment mutation in the tumour suppressor gene APC that otherwise prevents prolonged stabilisation of β-catenin. Surprisingly, no inhibitor compounds for WNT/β-catenin signalling have reached clinical use in part owing to the lack of specific in vivo assays that discriminate between on-target activities and dose-limiting toxicities. Here, we present a simple in vivo assay with a binary outcome whereby the administration of candidate compounds to pregnant and phenotypically normal Apcflox/flox mice can rescue in utero death of Apcmin/flox mutant conceptus without subsequent post-mortem assessment of WNT/β-catenin signalling. Indeed, the phenotypic plasticity of born Apcmin/flox conceptus enables future refinement of our assay to potentially enable dosage finding and cross-compound comparisons. Thus, we show for the first time the suitability of endogenous WNT/β-catenin signalling during embryonic development to provide an unambiguous and sensitive mammalian in vivo model to assess the efficacy and bioavailability of potential WNT/β-catenin antagonists.
Collapse
Affiliation(s)
- Janson Tse
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia (M.E.)
| | - Ryan O’Keefe
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia (M.E.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Angela Rigopolous
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia
| | - Annalisa L. E. Carli
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia (M.E.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Jo Waaler
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Rikshospitalet, 0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| | - Stefan Krauss
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Rikshospitalet, 0424 Oslo, Norway
- Hybrid Technology Hub-Centre of Excellence, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia (M.E.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - Michael Buchert
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia (M.E.)
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| |
Collapse
|
25
|
Khan S, Kwak YT, Peng L, Hu S, Cantarel BL, Lewis CM, Gao Y, Mani RS, Kanneganti TD, Zaki H. NLRP12 downregulates the Wnt/β-catenin pathway via interaction with STK38 to suppress colorectal cancer. J Clin Invest 2023; 133:e166295. [PMID: 37581937 PMCID: PMC10541192 DOI: 10.1172/jci166295] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/08/2023] [Indexed: 08/17/2023] Open
Abstract
Colorectal cancer (CRC) at advanced stages is rarely curable, underscoring the importance of exploring the mechanism of CRC progression and invasion. NOD-like receptor family member NLRP12 was shown to suppress colorectal tumorigenesis, but the precise mechanism was unknown. Here, we demonstrate that invasive adenocarcinoma development in Nlrp12-deficient mice is associated with elevated expression of genes involved in proliferation, matrix degradation, and epithelial-mesenchymal transition. Signaling pathway analysis revealed higher activation of the Wnt/β-catenin pathway, but not NF-κB and MAPK pathways, in the Nlrp12-deficient tumors. Using Nlrp12-conditional knockout mice, we revealed that NLRP12 downregulates β-catenin activation in intestinal epithelial cells, thereby suppressing colorectal tumorigenesis. Consistent with this, Nlrp12-deficient intestinal organoids and CRC cells showed increased proliferation, accompanied by higher activation of β-catenin in vitro. With proteomic studies, we identified STK38 as an interacting partner of NLRP12 involved in the inhibition of phosphorylation of GSK3β, leading to the degradation of β-catenin. Consistently, the expression of NLRP12 was significantly reduced, while p-GSK3β and β-catenin were upregulated in mouse and human colorectal tumor tissues. In summary, NLRP12 is a potent negative regulator of the Wnt/β-catenin pathway, and the NLRP12/STK38/GSK3β signaling axis could be a promising therapeutic target for CRC.
Collapse
Affiliation(s)
| | | | | | | | - Brandi L. Cantarel
- Bioinformatics Core Facility, Lyda Hill Department of Bioinformatics, and
| | - Cheryl M. Lewis
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | | | | | | |
Collapse
|
26
|
Li TH, Qin XH, Wang LQ, Qin C, Zhao BB, Cao HT, Yang XY, Wang YY, Li ZR, Zhou XT, Wang WB. Prognostic value and immune infiltration of ARMC10 in pancreatic adenocarcinoma via integrated bioinformatics analyses. Heliyon 2023; 9:e20464. [PMID: 37842592 PMCID: PMC10569960 DOI: 10.1016/j.heliyon.2023.e20464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 10/17/2023] Open
Abstract
Background Armadillo repeat-containing 10 (ARMC10) is involved in the progression of multiple types of tumors. Pancreatic adenocarcinoma (PAAD) is a lethal disease with poor survival and prognosis. Methods We acquired the data of ARMC10 in PAAD patients from the cancer genome atlas (TCGA) and gene expression omnibus (GEO) datasets and compared the expression level with normal pancreatic tissues. We evaluated the relevance between ARMC10 expression and clinicopathological factors, immune infiltration degree and prognosis in PAAD. Results High expression of ARMC10 was relevant to T stage, M stage, pathologic stage, histologic grade, residual tumor, primary therapy outcome (P < 0.05) and related to lower Overall-Survival (OS), Disease-Specific Survival (DSS), and Progression-Free Interval (PFI). Gene set enrichment analysis showed that ARMC10 was related to methylation in neural precursor cells (NPC), G alpha (i) signaling events, APC targets, energy metabolism, potassium channels and IL10 synthesis. The expression level of ARMC10 was positively related to the abundance of T helper cells and negatively to that of plasmacytoid dendritic cells (pDCs). Knocking down of ARMC10 could lead to lower proliferation, invasion, migration ability and colony formation rate of PAAD cells in vitro. Conclusions Our research firstly discovered ARMC10 as a novel prognostic biomarker for PAAD patients and played a crucial role in immune regulation in PAAD.
Collapse
Affiliation(s)
- Tian-Hao Li
- Department of Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiao-Han Qin
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Li-Quan Wang
- Department of Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bang-Bo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hong-Tao Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiao-Ying Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuan-Yang Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ze-Ru Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xing-Tong Zhou
- Department of Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wei-Bin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Li C, Furth EE, Rustgi AK, Klein PS. When You Come to a Fork in the Road, Take It: Wnt Signaling Activates Multiple Pathways through the APC/Axin/GSK-3 Complex. Cells 2023; 12:2256. [PMID: 37759479 PMCID: PMC10528086 DOI: 10.3390/cells12182256] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
The Wnt signaling pathway is a highly conserved regulator of metazoan development and stem cell maintenance. Activation of Wnt signaling is an early step in diverse malignancies. Work over the past four decades has defined a "canonical" Wnt pathway that is initiated by Wnt proteins, secreted glycoproteins that bind to a surface receptor complex and activate intracellular signal transduction by inhibiting a catalytic complex composed of the classical tumor suppressor Adenomatous Polyposis Coli (APC), Axin, and Glycogen Synthase Kinase-3 (GSK-3). The best characterized effector of this complex is β-catenin, which is stabilized by inhibition of GSK-3, allowing β-catenin entrance to the nucleus and activation of Wnt target gene transcription, leading to multiple cancers when inappropriately activated. However, canonical Wnt signaling through the APC/Axin/GSK-3 complex impinges on other effectors, independently of β-catenin, including the mechanistic Target of Rapamycin (mTOR), regulators of protein stability, mitotic spindle orientation, and Hippo signaling. This review focuses on these alternative effectors of the canonical Wnt pathway and how they may contribute to cancers.
Collapse
Affiliation(s)
- Chenchen Li
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Peter S. Klein
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
28
|
Yang X, Du Y, Luo L, Xu X, Xiong S, Yang X, Guo L, Liang T. Deciphering the Enigmatic Influence: Non-Coding RNAs Orchestrating Wnt/β-Catenin Signaling Pathway in Tumor Progression. Int J Mol Sci 2023; 24:13909. [PMID: 37762212 PMCID: PMC10530696 DOI: 10.3390/ijms241813909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Dysregulated expression of specific non-coding RNAs (ncRNAs) has been strongly linked to tumorigenesis, cancer progression, and therapeutic resistance. These ncRNAs can act as either oncogenes or tumor suppressors, thereby serving as valuable diagnostic and prognostic markers. Numerous studies have implicated the participation of ncRNAs in the regulation of diverse signaling pathways, including the pivotal Wnt/β-catenin signaling pathway that is widely acknowledged for its pivotal role in embryogenesis, cellular proliferation, and tumor biology control. Recent emerging evidence has shed light on the capacity of ncRNAs to interact with key components of the Wnt/β-catenin signaling pathway, thereby modulating the expression of Wnt target genes in cancer cells. Notably, the activity of this pathway can reciprocally influence the expression levels of ncRNAs. However, comprehensive analysis investigating the specific ncRNAs associated with the Wnt/β-catenin signaling pathway and their intricate interactions in cancer remains elusive. Based on these noteworthy findings, this review aims to unravel the intricate associations between ncRNAs and the Wnt/β-catenin signaling pathway during cancer initiation, progression, and their potential implications for therapeutic interventions. Additionally, we provide a comprehensive overview of the characteristics of ncRNAs and the Wnt/β-catenin signaling pathway, accompanied by a thorough discussion of their functional roles in tumor biology. Targeting ncRNAs and molecules associated with the Wnt/β-catenin signaling pathway may emerge as a promising and effective therapeutic strategy in future cancer treatments.
Collapse
Affiliation(s)
- Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Lulu Luo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Xinru Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| | - Shizheng Xiong
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Xueni Yang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China; (S.X.); (X.Y.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (X.Y.); (Y.D.); (L.L.); (X.X.)
| |
Collapse
|
29
|
Zhang X, Wu N, Huang H, Li S, Liu S, Zhang R, Huang Y, Lyu H, Xiao S, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Phosphorylated PTTG1 switches its subcellular distribution and promotes β-catenin stabilization and subsequent transcription activity. Oncogene 2023; 42:2439-2455. [PMID: 37400529 DOI: 10.1038/s41388-023-02767-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/05/2023]
Abstract
The Wnt/β-catenin signaling is usually abnormally activated in hepatocellular carcinoma (HCC), and pituitary tumor-transforming gene 1 (PTTG1) has been found to be highly expressed in HCC. However, the specific mechanism of PTTG1 pathogenesis remains poorly understood. Here, we found that PTTG1 is a bona fide β-catenin binding protein. PTTG1 positively regulates Wnt/β-catenin signaling by inhibiting the destruction complex assembly, promoting β-catenin stabilization and subsequent nuclear localization. Moreover, the subcellular distribution of PTTG1 was regulated by its phosphorylation status. Among them, PP2A induced PTTG1 dephosphorylation at Ser165/171 residues and prevented PTTG1 translocation into the nucleus, but these effects were effectively reversed by PP2A inhibitor okadaic acid (OA). Interestingly, we found that PTTG1 decreased Ser9 phosphorylation-inactivation of GSK3β by competitively binding to PP2A with GSK3β, indirectly leading to cytoplasmic β-catenin stabilization. Finally, PTTG1 was highly expressed in HCC and associated with poor patient prognosis. PTTG1 could promote the proliferative and metastasis of HCC cells. Overall, our results indicated that PTTG1 plays a crucial role in stabilizing β-catenin and facilitating its nuclear accumulation, leading to aberrant activation of Wnt/β-catenin signaling and providing a feasible therapeutic target for human HCC.
Collapse
Affiliation(s)
- Xuewen Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Nianping Wu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Huili Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shi Li
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shicheng Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G2R3, Canada.
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
30
|
Shin JY, Kim J, Choi YH, Lee S, Kang NG. Escin Activates Canonical Wnt/β-Catenin Signaling Pathway by Facilitating the Proteasomal Degradation of Glycogen Synthase Kinase-3β in Cultured Human Dermal Papilla Cells. Curr Issues Mol Biol 2023; 45:5902-5913. [PMID: 37504289 PMCID: PMC10377929 DOI: 10.3390/cimb45070373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/29/2023] Open
Abstract
Abnormal inactivation of the Wnt/β-catenin signaling pathway is involved in skin diseases like androgenetic alopecia, vitiligo and canities, but small-molecule activators are rarely described. In this study, we investigated the stimulatory effects of escin on the canonical Wnt/β-catenin signaling pathway in cultured human dermal papilla cells (hDPCs). Escin stimulated Wnt/β-catenin signaling, resulting in increased β-catenin and lymphoid enhancer-binding factor 1 (LEF1), the accumulation of nuclear β-catenin and the enhanced expression of Wnt target genes in cultured hDPCs. Escin drastically reduced the protein level of glycogen synthase kinase (GSK)-3β, a key regulator of the Wnt/β-catenin signaling pathway, while the presence of the proteasome inhibitor MG-132 fully restored the GSK-3β protein level. The treatment of secreted frizzled-related proteins (sFRPs) 1 and 2 attenuated the activity of escin in Wnt reporter assays. Our data demonstrate that escin is a natural agonist of the canonical Wnt/β-catenin signaling pathway and downregulates GSK-3β protein expression by facilitating the proteasomal degradation of GSK-3β in cultured hDPCs. Our data suggest that escin likely stimulates Wnt signaling through direct interactions with frizzled receptors. This study underscores the therapeutic potential of escin for Wnt-related diseases such as androgenetic alopecia, vitiligo and canities.
Collapse
Affiliation(s)
- Jae Young Shin
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul 07795, Republic of Korea
| | - Jaeyoon Kim
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul 07795, Republic of Korea
| | - Yun-Ho Choi
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul 07795, Republic of Korea
| | - Sanghwa Lee
- Innovo Therapeutics Inc., 507 38, Mapo-daero, Mapo-gu, Seoul 04174, Republic of Korea
| | - Nae-Gyu Kang
- LG Household & Health Care (LG H&H) R&D Center, 70, Magokjoongang 10-ro, Gangseo-gu, Seoul 07795, Republic of Korea
| |
Collapse
|
31
|
Zhou Y, Fyrner T, Chen CH, Sather NA, Hsu EL, Stupp SI, Snead ML. Optimization of peptide amphiphile-lipid raft interaction by changing peptide amphiphile lipophilicity. Acta Biomater 2023; 164:377-386. [PMID: 37040812 PMCID: PMC10225347 DOI: 10.1016/j.actbio.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/13/2023]
Abstract
Various peptide amphiphile (PA) molecules have been developed to promote bone regeneration. Previously we discovered that a peptide amphiphile with a palmitic acid tail (C16) attenuates the signaling threshold of leucine-rich amelogenin peptide (LRAP)-mediated Wnt activation by increasing membrane lipid raft mobility. In the current study, we found that treatment of murine ST2 cells with an inhibitor (Nystatin) or Caveolin-1-specific siRNA abolishes the effect of C16 PA, indicating that Caveolin-mediated endocytosis is required. To determine whether hydrophobicity of the PA tail plays a role in its signaling effect, we modified the length of the tail (C12, C16 and C22) or composition (cholesterol). While shortening the tail (C12) decreased the signaling effect, lengthening the tail (C22) had no prominent effect. On the other hand, the cholesterol PA displayed a similar function as the C16 PA at the same concentration of 0.001% w/v. Interestingly, a higher concentration of C16 PA (0.005%) is cytotoxic while cholesterol PA at the higher concentration (0.005%) is well-tolerated by cells. Use of the cholesterol PA at 0.005% enabled a further reduction of the signaling threshold of LRAP to 0.20 nM, compared to 0.25 nM at 0.001%. Caveolin-mediated endocytosis is also required for cholesterol PA, as evidenced by Caveolin-1 siRNA knockdown experiments. We further demonstrated that the noted effects of cholesterol PA are also observed in human bone marrow mesenchymal stem cells (BMMSCs). Taken together, these results indicate that the cholesterol PA modulates lipid raft/caveolar dynamics, thereby increasing receptor sensitivity for activation of canonical Wnt signaling. STATEMENT OF SIGNIFICANCE: Cell signaling involves not only the binding of growth factors (or other cytokines) and cognate receptors, but also their clustering on the cell membrane. However, little or no work has been directed thus far toward investigating how biomaterials can serve to enhance growth factor or peptide signaling by increasing diffusion of cell surface receptors within membrane lipid rafts. Therefore, a better understanding of the cellular and molecular mechanism(s) operating at the material-cell membrane interface during cell signaling has the potential to change the paradigm in designing future biomaterials and regenerative medicine therapeutics. In this study, we designed a peptide amphiphile (PA) with a cholesterol tail to enhance canonical Wnt signaling by modulating lipid raft/caveolar dynamics.
Collapse
Affiliation(s)
- Yan Zhou
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA.
| | - Timmy Fyrner
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Charlotte H Chen
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Nicholas A Sather
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Erin L Hsu
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Orthopaedic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Samuel I Stupp
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, USA; Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; Department of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Malcolm L Snead
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, 2250 Alcazar St, Los Angeles, CA 90033, USA
| |
Collapse
|
32
|
Tsai HP, Lin CJ, Lieu AS, Chen YT, Tseng TT, Kwan AL, Loh JK. Galectin-3 Mediates Tumor Progression in Astrocytoma by Regulating Glycogen Synthase Kinase-3β Activity. Curr Issues Mol Biol 2023; 45:3591-3602. [PMID: 37185758 PMCID: PMC10137203 DOI: 10.3390/cimb45040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Numerous studies have considered galectin-3 or Glycogen synthase kinase 3 beta (GSK3B) as a potential prognosis marker for various cancers. However, the correlation between the protein expression of galectin-3/GSK3B and the clinical parameters of astrocytoma has not been reported. This study aims to validate the correlation between the clinical outcomes and protein expression of galectin-3/GSK3B in astrocytoma. Immunohistochemistry staining was performed to detect galectin-3/GSK3B protein expression in patients with astrocytoma. The Chi-square test, Kaplan-Meier evaluation, and Cox regression analysis were used to determine the correlation between clinical parameters and galectin-3/GSK3B expression. Cell proliferation, invasion, and migration were compared between a non-siRNA group and a galectin-3/GSK3B siRNA group. Protein expression in galectin-3 or GSK3B siRNA-treated cells was evaluated using western blotting. Galectin-3 and GSK3B protein expression were significantly positively correlated with the World Health Organization (WHO) astrocytoma grade and overall survival time. Multivariate analysis revealed that WHO grade, galectin-3 expression, and GSK3B expression were independent prognostic factors for astrocytoma. Galectin-3 or GSK3B downregulation induced apoptosis and decreased cell numbers, migration, and invasion. siRNA-mediated gene silencing of galectin-3 resulted in the downregulation of Ki-67, cyclin D1, VEGF, GSK3B, p-GSK3B Ser9 (p-GSK3B S9), and β-catenin. In contrast, GSK3B knockdown only decreased Ki-67, VEGF, p-GSK3B S9, and β-catenin protein expression but did not affect cyclin D1 and galectin-3 protein expression. The siRNA results indicated that GSK3B is downstream of the galectin-3 gene. These data support that galectin-3 mediated tumor progression by upregulating GSK3B and β-catenin protein expression in glioblastoma. Therefore, galectin-3 and GSK3B are potential prognostic markers, and their genes may be considered to be anticancer targets for astrocytoma therapy.
Collapse
Affiliation(s)
- Hung-Pei Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Chien-Ju Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Tzu-Ting Tseng
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22903, USA
| | - Joon-Khim Loh
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
33
|
Morita M, Nishida N, Aoki T, Chishina H, Takita M, Ida H, Hagiwara S, Minami Y, Ueshima K, Kudo M. Role of β-Catenin Activation in the Tumor Immune Microenvironment and Immunotherapy of Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:cancers15082311. [PMID: 37190239 DOI: 10.3390/cancers15082311] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Recently, the therapeutic combination of atezolizumab and bevacizumab was widely used to treat advanced hepatocellular carcinoma (HCC). According to recent clinical trials, immune checkpoint inhibitors (ICIs) and molecular target agents are expected to be key therapeutic strategies in the future. Nonetheless, the mechanisms underlying molecular immune responses and immune evasion remain unclear. The tumor immune microenvironment plays a vital role in HCC progression. The infiltration of CD8-positive cells into tumors and the expression of immune checkpoint molecules are key factors in this immune microenvironment. Specifically, Wnt/β catenin pathway activation causes "immune exclusion", associated with poor infiltration of CD8-positive cells. Some clinical studies suggested an association between ICI resistance and β-catenin activation in HCC. Additionally, several subclassifications of the tumor immune microenvironment were proposed. The HCC immune microenvironment can be broadly divided into inflamed class and non-inflamed class, with several subclasses. β-catenin mutations are important factors in immune subclasses; this may be useful when considering therapeutic strategies as β-catenin activation may serve as a biomarker for ICI. Various types of β-catenin modulators were developed. Several kinases may also be involved in the β-catenin pathway. Therefore, combinations of β-catenin modulators, kinase inhibitors, and ICIs may exert synergistic effects.
Collapse
Affiliation(s)
- Masahiro Morita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Tomoko Aoki
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Hirokazu Chishina
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Masahiro Takita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Hiroshi Ida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Satoru Hagiwara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Kazuomi Ueshima
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama 589-8511, Japan
| |
Collapse
|
34
|
Pećina-Šlaus N, Aničić S, Bukovac A, Kafka A. Wnt Signaling Inhibitors and Their Promising Role in Tumor Treatment. Int J Mol Sci 2023; 24:ijms24076733. [PMID: 37047705 PMCID: PMC10095594 DOI: 10.3390/ijms24076733] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
In a continuous search for the improvement of antitumor therapies, the inhibition of the Wnt signaling pathway has been recognized as a promising target. The altered functioning of the Wnt signaling in human tumors points to the strategy of the inhibition of its activity that would impact the clinical outcomes and survival of patients. Because the Wnt pathway is often mutated or epigenetically altered in tumors, which promotes its activation, inhibitors of Wnt signaling are being intensively investigated. It has been shown that knocking down specific components of the Wnt pathway has inhibitory effects on tumor growth in vivo and in vitro. Thus, similar effects are expected from the application of Wnt inhibitors. In the last decades, molecules acting as inhibitors on the pathway’s specific molecular levels have been identified and characterized. This review will discuss the inhibitors of the canonical Wnt pathway, summarize knowledge on their effectiveness as therapeutics, and debate their side effects. The role of the components frequently mutated in various tumors that are principal targets for Wnt inhibitors is also going to be brought to the reader’s attention. Some of the molecules identified as Wnt pathway inhibitors have reached early stages of clinical trials, and some have only just been discovered. All things considered, inhibition of the Wnt signaling pathway shows potential for the development of future therapies.
Collapse
Affiliation(s)
- Nives Pećina-Šlaus
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| | - Sara Aničić
- Department of Physiology and Immunology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Anja Bukovac
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| | - Anja Kafka
- Laboratory of Neuro-Oncology, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Šalata 12, 10000 Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Šalata 3, 10000 Zagreb, Croatia
| |
Collapse
|
35
|
Bressler SG, Mitrany A, Wenger A, Näthke I, Friedler A. The Oligomerization Domains of the APC Protein Mediate Liquid-Liquid Phase Separation That Is Phosphorylation Controlled. Int J Mol Sci 2023; 24:ijms24076478. [PMID: 37047451 PMCID: PMC10095272 DOI: 10.3390/ijms24076478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
One of the most important properties of intrinsically disordered proteins is their ability to undergo liquid-liquid phase separation and form droplets. The Adenomatous Polyposis Coli (APC) protein is an IDP that plays a key role in Wnt signaling and mutations in Apc initiate cancer. APC forms droplets via its 20R domains and self-association domain (ASAD) and in the context of Axin. However, the mechanism involved is unknown. Here, we used peptides to study the molecular mechanism and regulation of APC droplet formation. We found that a peptide derived from the ASAD of APC-formed droplets. Peptide array screening showed that the ASAD bound other APC peptides corresponding to the 20R3 and 20R5 domains. We discovered that the 20R3/5 peptides also formed droplets by themselves and mapped specific residues within 20R3/5 that are necessary for droplet formation. When incubated together, the ASAD and 20R3/5 did not form droplets. Thus, the interaction of the ASAD with 20R3 and 20R5 may regulate the droplet formation as a means of regulating different cellular functions. Phosphorylation of 20R3 or 20R5 at specific residues prevented droplet formation of 20R3/5. Our results reveal that phosphorylation and the ability to undergo liquid-liquid phase separation, which are both important properties of intrinsically disordered proteins, are related to each other in APC. Phosphorylation inhibited the liquid-liquid phase separation of APC, acting as an ‘on-off’ switch for droplet formation. Phosphorylation may thus be a common mechanism regulating LLPS in intrinsically disordered proteins.
Collapse
Affiliation(s)
- Shachar G. Bressler
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Amit Mitrany
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Alon Wenger
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Inke Näthke
- Division of Molecular Cell and Developmental Biology, University of Dundee, Dundee DD1 5AA, Scotland, UK
- Correspondence: (I.N.); (A.F.)
| | - Assaf Friedler
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel
- Correspondence: (I.N.); (A.F.)
| |
Collapse
|
36
|
Yakkala P, Panda SR, Naidu VGM, Shafi S, Kamal A. Pyridine-Based 1,2,4-Triazolo-Tethered Indole Conjugates Potentially Affecting TNKS and PI3K in Colorectal Cancer. ACS Med Chem Lett 2023; 14:260-269. [PMID: 36923920 PMCID: PMC10009797 DOI: 10.1021/acsmedchemlett.2c00475] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
A library of pyridine-based 1,2,4-triazolo-tethered indole conjugates were designed, synthesized, and evaluated for anti-proliferative activity against a panel of six human cancer cell lines. All the synthesized conjugates (14a-q) were found to be effective against the HT-29 cell line. Particularly conjugates 14a, 14n, and 14q exhibited promising cytotoxicity, with IC50 values of 1 μM, 2.4 μM, and 3.6 μM, respectively, compared to the standard 5-fluorouracil (IC50 = 5.31 μM). Cell cycle arrest at the G0/G1 phase was observed with these compounds, the mitochondrial membrane potential was interrupted, and the total ROS production was enhanced. Western blot and immunofluorescence experiments illustrated that these compounds inhibit the expression of markers that are involved in β-catenin and PI3K pathways. Molecular dynamics simulations demonstrated that compound 14a has major hydrophobic interactions and few H-bonding interactions with both PI3K and tankyrase proteins.
Collapse
Affiliation(s)
- Prasanna
A. Yakkala
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Education and
Research, Jamia Hamdard, New Delhi 110062, India
| | - Samir R. Panda
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India
| | - Vegi G. M. Naidu
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India
| | - Syed Shafi
- Department
of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Ahmed Kamal
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Education and
Research, Jamia Hamdard, New Delhi 110062, India
- Department
of Pharmacy, Birla Institute of Technology
& Science, Pilani, Hyderabad Campus, Hyderabad 500078, TS, India
| |
Collapse
|
37
|
Reprogramming of palmitic acid induced by dephosphorylation of ACOX1 promotes β-catenin palmitoylation to drive colorectal cancer progression. Cell Discov 2023; 9:26. [PMID: 36878899 PMCID: PMC9988979 DOI: 10.1038/s41421-022-00515-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 12/30/2022] [Indexed: 03/08/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer. However, it is not well known how metabolism affects cancer progression. We identified that metabolic enzyme acyl-CoA oxidase 1 (ACOX1) suppresses colorectal cancer (CRC) progression by regulating palmitic acid (PA) reprogramming. ACOX1 is highly downregulated in CRC, which predicts poor clinical outcome in CRC patients. Functionally, ACOX1 depletion promotes CRC cell proliferation in vitro and colorectal tumorigenesis in mouse models, whereas ACOX1 overexpression inhibits patient-derived xenograft growth. Mechanistically, DUSP14 dephosphorylates ACOX1 at serine 26, promoting its polyubiquitination and proteasomal degradation, thereby leading to an increase of the ACOX1 substrate PA. Accumulated PA promotes β-catenin cysteine 466 palmitoylation, which inhibits CK1- and GSK3-directed phosphorylation of β-catenin and subsequent β-Trcp-mediated proteasomal degradation. In return, stabilized β-catenin directly represses ACOX1 transcription and indirectly activates DUSP14 transcription by upregulating c-Myc, a typical target of β-catenin. Finally, we confirmed that the DUSP14-ACOX1-PA-β-catenin axis is dysregulated in clinical CRC samples. Together, these results identify ACOX1 as a tumor suppressor, the downregulation of which increases PA-mediated β-catenin palmitoylation and stabilization and hyperactivates β-catenin signaling thus promoting CRC progression. Particularly, targeting β-catenin palmitoylation by 2-bromopalmitate (2-BP) can efficiently inhibit β-catenin-dependent tumor growth in vivo, and pharmacological inhibition of DUSP14-ACOX1-β-catenin axis by Nu-7441 reduced the viability of CRC cells. Our results reveal an unexpected role of PA reprogramming induced by dephosphorylation of ACOX1 in activating β-catenin signaling and promoting cancer progression, and propose the inhibition of the dephosphorylation of ACOX1 by DUSP14 or β-catenin palmitoylation as a viable option for CRC treatment.
Collapse
|
38
|
Electrochemical biosensors for analysis of DNA point mutations in cancer research. Anal Bioanal Chem 2023; 415:1065-1085. [PMID: 36289102 DOI: 10.1007/s00216-022-04388-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 02/07/2023]
Abstract
Cancer is a genetic disease induced by mutations in DNA, in particular point mutations in important driver genes that lead to protein malfunctioning and ultimately to tumorigenesis. Screening for the most common DNA point mutations, especially in such genes as TP53, BRCA1 and BRCA2, EGFR, KRAS, or BRAF, is crucial to determine predisposition risk for cancer or to predict response to therapy. In this review, we briefly depict how these genes are involved in cancer, followed by a description of the most common techniques routinely applied for their analysis, including high-throughput next-generation sequencing technology and less expensive low-throughput options, such as real-time PCR, restriction fragment length polymorphism, or high resolution melting analysis. We then introduce benefits of electrochemical biosensors as interesting alternatives to the standard methods in terms of cost, speed, and simplicity. We describe most common strategies involved in electrochemical biosensing of point mutations, relying mostly on PCR or isothermal amplification techniques, and critically discuss major challenges and obstacles that, until now, prevented their more widespread application in clinical settings.
Collapse
|
39
|
Gu H, Wei J. Peiminine regulates bone-fat balance by canonical Wnt/β-catenin pathway in an ovariectomized rat model. Phytother Res 2023. [PMID: 36799485 DOI: 10.1002/ptr.7780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 11/19/2022] [Accepted: 01/29/2023] [Indexed: 02/18/2023]
Abstract
Peiminine is a major biologically active component of Fritillaria thunbergii Miq that exhibits good anticancer, antiinflammatory, and anti-osteoclast effects. However, its effects on osteoporosis (OP) remain unknown. This study aimed to explore whether Peiminine was able to regulate osteogenesis and adipogenesis in ovariectomized (OVX) rat. The effects on the differentiation of bone marrow stem cells (BMSCs), function of Wnt/β-catenin pathway, ALP activity, calcium nodule deposition, as well as adipocyte formation in vitro by Peiminine at different concentrations, were detected. The curative effects of Peiminine on the ovariectomy-induced osteoporosis model by micro-CT and bone histomorphology assays were analyzed. The promotion of osteogenic differentiation and inhibition of adipogenic differentiation by Peiminine (5-40 μg/mL) was detected and the optimum concentration was 20 μg/mL. Mechanistically, Peiminine regulated the fate of BMSCs in vitro, and activated Wnt/β-catenin signaling pathway by restraining phosphorylation of β-catenin and promoting the nuclear translocation of β-catenin. Moreover, Peiminine prevented ovariectomy-induced osteoporosis by alleviating trabecular bone loss and inhibiting adipose formation. Our data suggested that Peiminine could attenuate ovariectomy-induced osteoporosis by alleviating trabecular bone loss and inhibiting adipose formation. These encouraging discoveries could lay the foundation for Peiminine to be a promising preventive treatment strategy for skeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Hanwen Gu
- Department of Joint Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jian Wei
- Department of Joint Orthopedics, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, China
| |
Collapse
|
40
|
Fernandes B, Cavaco-Paulo A, Matamá T. A Comprehensive Review of Mammalian Pigmentation: Paving the Way for Innovative Hair Colour-Changing Cosmetics. BIOLOGY 2023; 12:biology12020290. [PMID: 36829566 PMCID: PMC9953601 DOI: 10.3390/biology12020290] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/26/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
The natural colour of hair shafts is formed at the bulb of hair follicles, and it is coupled to the hair growth cycle. Three critical processes must happen for efficient pigmentation: (1) melanosome biogenesis in neural crest-derived melanocytes, (2) the biochemical synthesis of melanins (melanogenesis) inside melanosomes, and (3) the transfer of melanin granules to surrounding pre-cortical keratinocytes for their incorporation into nascent hair fibres. All these steps are under complex genetic control. The array of natural hair colour shades are ascribed to polymorphisms in several pigmentary genes. A myriad of factors acting via autocrine, paracrine, and endocrine mechanisms also contributes for hair colour diversity. Given the enormous social and cosmetic importance attributed to hair colour, hair dyeing is today a common practice. Nonetheless, the adverse effects of the long-term usage of such cosmetic procedures demand the development of new methods for colour change. In this context, case reports of hair lightening, darkening and repigmentation as a side-effect of the therapeutic usage of many drugs substantiate the possibility to tune hair colour by interfering with the biology of follicular pigmentary units. By scrutinizing mammalian pigmentation, this review pinpoints key targetable processes for the development of innovative cosmetics that can safely change the hair colour from the inside out.
Collapse
Affiliation(s)
- Bruno Fernandes
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Artur Cavaco-Paulo
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (A.C.-P.); (T.M.); Tel.: +351-253-604-409 (A.C.-P.); +351-253-601-599 (T.M.)
| | - Teresa Matamá
- CEB—Centre of Biological Engineering, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- LABBELS—Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (A.C.-P.); (T.M.); Tel.: +351-253-604-409 (A.C.-P.); +351-253-601-599 (T.M.)
| |
Collapse
|
41
|
3,3'-Diindolylmethane Augments 5-Fluorouracil-InducedGrowth Suppression in Gastric Cancer Cells through Suppression of the Akt/GSK-3 β and WNT/Beta-Catenin. JOURNAL OF ONCOLOGY 2023; 2023:8268955. [PMID: 36785670 PMCID: PMC9922186 DOI: 10.1155/2023/8268955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 02/09/2023]
Abstract
Gastric cancer (GC) is one of the most lethal cancers in South Korea, and it is a cancer of concern worldwide. 5-fluorouracil (5-Fu) is commonly used as the first-line therapy for advanced GC; however, its side effects often limit the dosage range and impair patients' quality of life. Due to the limitations of current chemotherapy, new anticancer therapies are urgently needed. 3,3'-diindolylmethane (DIM) has been reported to have the ability to protect against various types of cancer. Our study aimed to elucidate the anticancer effect of DIM in GC when treated with the chemotherapeutic agent 5-Fu. In our results, combined treatment with DIM and 5-Fu resulted in higher apoptosis and lower cell proliferation than treatment with 5-Fu in SNU484 and SNU638 cell lines. Furthermore, when DIM and 5-Fu were administered together, cell invasion was diminished by mediated E-cadherin, MMP-9, and uPA; p-Akt and p-GSK-3β levels were reduced more significantly than when 5-Fu was administered alone. Moreover, in the Wnt signaling pathway, combined treatment of DIM and 5-Fu diminished β-catenin levels in the nucleus and inhibited cyclin D1and c-Myc protein levels. The Akt inhibitor, wortmannin, further inhibited the levels of β-catenin and c-Myc that were inhibited by DIM and 5-Fu. Furthermore, an animal xenograft model demonstrated that DIM combined with 5-Fu considerably reduced tumor growth without any toxic effects by regulating the Akt/GSK-3β and β-catenin levels. Our findings suggest that DIM significantly potentiates the anticancer effects of 5-Fu by targeting the Akt/GSK-3β and WNT/β-catenin because the combination therapy is more effective than 5-Fu alone, thereby offering an innovative potential therapy for patients with GC.
Collapse
|
42
|
Kim DY, Ryu YS, Lee ES, Koh DI, Moon JH, Jung SA, Kim MJ, Yun H, You JE, Jeong HR, Yoon DI, Kim CH, Hong SW, Gong YD, Jin DH. DGG-300273, a novel WNT/β-catenin inhibitor, induces apoptotic cell death by activating ROS-BIM signaling in a Wnt-dependent manner in colon cancer cells. Invest New Drugs 2023; 41:105-114. [PMID: 36538258 DOI: 10.1007/s10637-022-01295-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022]
Abstract
Dysregulated Wnt signaling is associated with malignant oncogenic transformation, especially in colon cancer. Recently, numerous drugs have been developed based on tumorigenesis biomarkers, thus having high potential as drug targets. Likewise, WNT/β-catenin pathway members are attractive therapeutic targets for colon cancer and are currently in various stages of development. However, although inhibitors of proteins regulating the WNT/β-catenin signaling pathway have been extensively studied, they have yet to be clinically approved, and the underlying molecular mechanism(s) of their anticancer effects remain poorly understood. Herein, we show that a novel WNT/β-catenin inhibitor, DGG-300273, inhibits colon cancer cell growth in a Wnt-dependent manner due to upregulation of the BCL2-family protein Bim and caspase-dependent apoptotic cell death. Additionally, DGG-300273-mediated cell death occurs by increased reactive oxygen species (ROS), as shown by abrogation of apoptotic cell death and ROS production following pretreatment with the antioxidant N-acetylcysteine. These results suggest that DGG-300273 represents a promising investigational drug for the treatment of Wnt-associated cancer, thus warranting further characterization and study.
Collapse
Affiliation(s)
- Do Yeon Kim
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Yea Seong Ryu
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Eun-Sil Lee
- Innovative Drug-Like Library Research Center, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul, 04620, Republic of Korea
| | - Dong-In Koh
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jai-Hee Moon
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Soo-A Jung
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Mi Jin Kim
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hyeseon Yun
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Ji-Eun You
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Hong-Rae Jeong
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Dong-Il Yoon
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Chul Hee Kim
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Seung-Woo Hong
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Young-Dae Gong
- Innovative Drug-Like Library Research Center, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul, 04620, Republic of Korea.
| | - Dong-Hoon Jin
- Asan Institute for Life Science, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, 05505, Republic of Korea.
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympicro-43 gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
43
|
Fang Z, Meng Q, Xu J, Wang W, Zhang B, Liu J, Liang C, Hua J, Zhao Y, Yu X, Shi S. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun (Lond) 2023; 43:3-41. [PMID: 36424360 PMCID: PMC9859735 DOI: 10.1002/cac2.12392] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022] Open
Abstract
As a critical component of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) play important roles in cancer initiation and progression. Well-known signaling pathways, including the transforming growth factor-β (TGF-β), Hedgehog (Hh), Notch, Wnt, Hippo, nuclear factor kappa-B (NF-κB), Janus kinase (JAK)/signal transducer and activator of transcription (STAT), mitogen-activated protein kinase (MAPK), and phosphoinositide 3-kinase (PI3K)/AKT pathways, as well as transcription factors, including hypoxia-inducible factor (HIF), heat shock transcription factor 1 (HSF1), P53, Snail, and Twist, constitute complex regulatory networks in the TME to modulate the formation, activation, heterogeneity, metabolic characteristics and malignant phenotype of CAFs. Activated CAFs remodel the TME and influence the malignant biological processes of cancer cells by altering the transcriptional and secretory characteristics, and this modulation partially depends on the regulation of signaling cascades. The results of preclinical and clinical trials indicated that therapies targeting signaling pathways in CAFs demonstrated promising efficacy but were also accompanied by some failures (e.g., NCT01130142 and NCT01064622). Hence, a comprehensive understanding of the signaling cascades in CAFs might help us better understand the roles of CAFs and the TME in cancer progression and may facilitate the development of more efficient and safer stroma-targeted cancer therapies. Here, we review recent advances in studies of signaling pathways in CAFs and briefly discuss some future perspectives on CAF research.
Collapse
Affiliation(s)
- Zengli Fang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Qingcai Meng
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jin Xu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Wei Wang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Bo Zhang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jiang Liu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Chen Liang
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jie Hua
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Yingjun Zhao
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Xianjun Yu
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Si Shi
- Department of Pancreatic SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
- Shanghai Pancreatic Cancer InstituteShanghai200032P. R. China
- Pancreatic Cancer InstituteFudan UniversityShanghai200032P. R. China
| |
Collapse
|
44
|
Zou G, Park JI. Wnt signaling in liver regeneration, disease, and cancer. Clin Mol Hepatol 2023; 29:33-50. [PMID: 35785913 PMCID: PMC9845677 DOI: 10.3350/cmh.2022.0058] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/30/2022] [Indexed: 02/02/2023] Open
Abstract
The liver exhibits the highest recovery rate from acute injuries. However, in chronic liver disease, the long-term loss of hepatocytes often leads to adverse consequences such as fibrosis, cirrhosis, and liver cancer. The Wnt signaling plays a pivotal role in both liver regeneration and tumorigenesis. Therefore, manipulating the Wnt signaling has become an attractive approach to treating liver disease, including cancer. Nonetheless, given the crucial roles of Wnt signaling in physiological processes, blocking Wnt signaling can also cause several adverse effects. Recent studies have identified cancer-specific regulators of Wnt signaling, which would overcome the limitation of Wnt signaling target approaches. In this review, we discussed the role of Wnt signaling in liver regeneration, precancerous lesion, and liver cancer. Furthermore, we summarized the basic and clinical approaches of Wnt signaling blockade and proposed the therapeutic prospects of cancer-specific Wnt signaling blockade for liver cancer treatment.
Collapse
Affiliation(s)
- Gengyi Zou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Corresponding author : Gengyi Zou Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd Unit 1054, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA,Jae-Il Park Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd. Unit 1052, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| |
Collapse
|
45
|
Al-Thani NM, Schaefer-Ramadan S, Aleksic J, Mohamoud YA, Malek JA. Identifying novel interactions of the colon-cancer related APC protein with Wnt-pathway nuclear transcription factors. Cancer Cell Int 2022; 22:376. [PMID: 36457029 PMCID: PMC9714242 DOI: 10.1186/s12935-022-02799-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/19/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Colon cancer is often driven by mutations of the adenomatous polyposis coli (APC) gene, an essential tumor suppressor gene of the Wnt β-catenin signaling pathway. APC and its cytoplasmic interactions have been well studied. However, various groups have also observed its presence in the nucleus. Identifying novel interactions of APC in the Wnt pathway will provide an opportunity to understand APC's nuclear role better and ultimately identify potential cancer treatment targets. METHODS We used the all-vs-all sequencing (AVA-Seq) method to interrogate the interactome of protein fragments spanning most of the 60 Wnt β-catenin pathway proteins. Using protein fragments identified the interacting regions between the proteins with more resolution than a full-length protein approach. Pull-down assays were used to validate a subset of these interactions. RESULTS 74 known and 703 novel Wnt β-catenin pathway protein-protein interactions were recovered in this study. There were 8 known and 31 novel APC protein-protein interactions. Novel interactions of APC and nuclear transcription factors TCF7, JUN, FOSL1, and SOX17 were particularly interesting and confirmed in validation assays. CONCLUSION Based on our findings of novel interactions between APC and transcription factors and previous evidence of APC localizing to the nucleus, we suggest APC may compete and repress CTNNB1. This would occur through APC binding to the transcription factors (JUN, FOSL1, TCF7) to regulate the Wnt signaling pathway including through enhanced marking of CTNNB1 for degradation in the nucleus by APC binding with SOX17. Additional novel Wnt β-catenin pathway protein-protein interactions from this study could lead researchers to novel drug designs for cancer.
Collapse
Affiliation(s)
- Nayra M. Al-Thani
- grid.416973.e0000 0004 0582 4340Department of Genetic Medicine, Weill Cornell Medicine in Qatar, PO Box 24144, Doha, Qatar ,grid.452146.00000 0004 1789 3191Department of Genomics and Precision Medicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Stephanie Schaefer-Ramadan
- grid.416973.e0000 0004 0582 4340Department of Genetic Medicine, Weill Cornell Medicine in Qatar, PO Box 24144, Doha, Qatar
| | - Jovana Aleksic
- grid.416973.e0000 0004 0582 4340Department of Genetic Medicine, Weill Cornell Medicine in Qatar, PO Box 24144, Doha, Qatar
| | - Yasmin A. Mohamoud
- grid.416973.e0000 0004 0582 4340Genomics Core, Weill Cornell Medicine in Qatar, Doha, Qatar
| | - Joel A. Malek
- grid.416973.e0000 0004 0582 4340Department of Genetic Medicine, Weill Cornell Medicine in Qatar, PO Box 24144, Doha, Qatar ,grid.416973.e0000 0004 0582 4340Genomics Core, Weill Cornell Medicine in Qatar, Doha, Qatar
| |
Collapse
|
46
|
Sileo P, Simonin C, Melnyk P, Chartier-Harlin MC, Cotelle P. Crosstalk between the Hippo Pathway and the Wnt Pathway in Huntington's Disease and Other Neurodegenerative Disorders. Cells 2022; 11:cells11223631. [PMID: 36429058 PMCID: PMC9688160 DOI: 10.3390/cells11223631] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
The Hippo pathway consists of a cascade of kinases that controls the phosphorylation of the co-activators YAP/TAZ. When unphosphorylated, YAP and TAZ translocate into the nucleus, where they mainly bind to the TEAD transcription factor family and activate genes related to cell proliferation and survival. In this way, the inhibition of the Hippo pathway promotes cell survival, proliferation, and stemness fate. Another pathway can modulate these processes, namely the Wnt/β-catenin pathway that is indeed involved in cellular functions such as proliferation and cell survival, as well as apoptosis, growth, and cell renewal. Wnt signaling can act in a canonical or noncanonical way, depending on whether β-catenin is involved in the process. In this review, we will focus only on the canonical Wnt pathway. It has emerged that YAP/TAZ are components of the β-catenin destruction complex and that there is a close relationship between the Hippo pathway and the canonical Wnt pathway. Furthermore, recent data have shown that both of these pathways may play a role in neurodegenerative diseases, such as Huntington's disease, Alzheimer's disease, or Amyotrophic Lateral Sclerosis. Thus, this review analyzes the Hippo pathway and the Wnt pathway, their crosstalk, and their involvement in Huntington's disease, as well as in other neurodegenerative disorders. Altogether, these data suggest possible therapeutic approaches targeting key players of these pathways.
Collapse
Affiliation(s)
- Pasquale Sileo
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
| | - Clémence Simonin
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- Centre de Référence Maladie de Huntington, CHU Lille, F-59000 Lille, France
| | - Patricia Melnyk
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
| | - Marie-Christine Chartier-Harlin
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- Correspondence: (M.-C.C.-H.); (P.C.)
| | - Philippe Cotelle
- Univ. Lille, INSERM, CHU Lille, UMR-S 1172, Lille Neuroscience and Cognition Research Center, F-59000 Lille, France
- ENSCL-Centrale Lille, CS 90108, F-59652 Villeneuve d’Ascq, France
- Correspondence: (M.-C.C.-H.); (P.C.)
| |
Collapse
|
47
|
Nalli M, Masci D, Urbani A, La Regina G, Silvestri R. Emerging Direct Targeting β-Catenin Agents. Molecules 2022; 27:molecules27227735. [PMID: 36431838 PMCID: PMC9698307 DOI: 10.3390/molecules27227735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Aberrant accumulation of β-catenin in the cell nucleus as a result of deregulation of the Wnt/β-catenin pathway is found in various types of cancer. Direct β-catenin targeting agents are being researched despite obstacles; however, specific β-catenin drugs for clinical treatments have not been approved so far. We focused on direct β-catenin targeting of potential therapeutic value as anticancer agents. This review provides recent advances on small molecule β-catenin agents. Structure-activity relationships and biological activities of reported inhibitors are discussed. This work provides useful knowledge in the discovery of β-catenin agents.
Collapse
Affiliation(s)
- Marianna Nalli
- Laboratory Affiliated with the Institute Pasteur Italy—Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated with the Institute Pasteur Italy—Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Romano Silvestri
- Laboratory Affiliated with the Institute Pasteur Italy—Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
48
|
Luo F, Li J, Liu J, Liu K. Stabilizing and upregulating Axin with tankyrase inhibitor reverses 5-fluorouracil chemoresistance and proliferation by targeting the WNT/caveolin-1 axis in colorectal cancer cells. Cancer Gene Ther 2022; 29:1707-1719. [PMID: 35750753 DOI: 10.1038/s41417-022-00493-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 05/11/2022] [Accepted: 06/08/2022] [Indexed: 02/04/2023]
Abstract
Chemoresistance is a main obstacle for colorectal cancer treatment. In this study, we evaluated the effects and mechanisms of the WNT/β-catenin signaling pathway on the chemoresistance of SW480 and SW620 colorectal cancer cells. The activity of β-catenin was activated/inhibited by the small molecule compound GSK-3 inhibitor 6-bromo-indirubin-3'-oxime and the tankyrase inhibitor XAV939. The downstream target genes of the WNT/β-catenin signaling pathway were screened using a cDNA microarray and bioinformatics analysis. Apoptosis induced by 5-Fu, cell cycle distribution and expression levels of WNT/β-catenin/TCF12/caveolin-1 and multidrug resistance proteins were examed by flow cytometry and western blot after β-catenin activation/inhibition and caveolin-1 overexpression/interference. The effect and mechanism of XAV939 on proliferation and apoptosis induced by 5-Fu in xenograft tumors of nude mice were evaluated by immunohistochemistry and TUNEL staining. 6-Bromo-indirubin-3'-oxime treatment increased β-catenin expression by regulating GSK-3β phosphorylation, accompanied by upregulation of TCF12, caveolin-1, P-gp, and MRP2 and downregulation of apoptosis induced by 5-Fu. Conversely, XAV939 treatment decreased β-catenin expression by upregulating Axin, accompanied by downregulation of TCF12, Caveolin-1, P-gp, and MRP2 and upregulation of apoptosis induced by 5-Fu. The caveolin-1 gene was identified as an important downstream gene of the WNT/β-catenin signaling pathway. Caveolin-1 overexpression upregulated β-catenin expression, increased P-gp and MRP2 expression and decreased apoptosis induced by 5-Fu; conversely, caveolin-1 interference caused the opposite effects. In addition, in vivo experiments showed that XAV939 treatment reduced β-catenin expression, increased apoptosis induced by 5-Fu and repressed xenograft tumor growth. Our findings suggested that inhibition of WNT/β-catenin/TCF12/caveolin-1 provides a new promising therapeutic strategy for colorectal cancer treatment.
Collapse
Affiliation(s)
- Feng Luo
- Department of Pathology, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511518, PR China
| | - Jinbang Li
- Department of Pathology, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511518, PR China
| | - Jihong Liu
- Department of Pathology, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511518, PR China
| | - Kunping Liu
- Department of Pathology, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, 511518, PR China.
| |
Collapse
|
49
|
Rai A, Yelamanchi SD, Radotra BD, Gupta SK, Mukherjee KK, Tripathi M, Chhabra R, Ahuja CK, Kumar N, Pandey A, Korbonits M, Dutta P, Gaston-Massuet C. Phosphorylation of β-catenin at Serine552 correlates with invasion and recurrence of non-functioning pituitary neuroendocrine tumours. Acta Neuropathol Commun 2022; 10:138. [PMID: 36114575 PMCID: PMC9482208 DOI: 10.1186/s40478-022-01441-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Non-functioning pituitary tumours (NF-PitNETs) are common intracranial benign neoplasms that can exhibit aggressive behaviour by invading neighbouring structures and, in some cases, have multiple recurrences. Despite resulting in severe co-morbidities, no predictive biomarkers of recurrence have been identified for NF-PitNETs. In this study we have used high-throughput mass spectrometry-based analysis to examine the phosphorylation pattern of different subsets of NF-PitNETs. Based on histopathological, radiological, surgical and clinical features, we have grouped NF-PitNETs into non-invasive, invasive, and recurrent disease groups. Tumour recurrence was determined based on regular clinical and radiological data of patients for a mean follow-up of 10 years (SD ± 5.4 years). Phosphoproteomic analyses identified a unique phosphopeptide enrichment pattern which correlates with disease recurrence. Candidate phosphorylated proteins were validated in a large cohort of NF-PitNET patients by western blot and immunohistochemistry. We identified a cluster of 22 phosphopeptides upregulated in recurrent NF-PitNETs compared to non-invasive and invasive subgroups. We reveal significant phosphorylation of the β-catenin at Ser552 in recurrent and invasive NF-PitNETs, compared to non-invasive/non-recurrent NF-PitNET subgroup. Moreover, β-catenin pSer552 correlates with the recurrence free survival among 200 patients with NF-PitNET. Together, our results suggest that the phosphorylation status of β-catenin at Ser552 could act as potential biomarker of tumour recurrence in NF-PitNETs.
Collapse
|
50
|
Proinflammatory and Cancer-Promoting Pathobiont Fusobacterium nucleatum Directly Targets Colorectal Cancer Stem Cells. Biomolecules 2022; 12:biom12091256. [PMID: 36139097 PMCID: PMC9496236 DOI: 10.3390/biom12091256] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Intestinal bacterial communities participate in gut homeostasis and are recognized as crucial in bowel inflammation and colorectal cancer (CRC). Fusobacterium nucleatum (Fn), a pathobiont of the oral microflora, has recently emerged as a CRC-associated microbe linked to disease progression, metastasis, and a poor clinical outcome; however, the primary cellular and/or microenvironmental targets of this agent remain elusive. We report here that Fn directly targets putative colorectal cancer stem cells (CR-CSCs), a tumor cell subset endowed with cancer re-initiating capacity after surgery and chemotherapy. A patient-derived CSC line, highly enriched (70%) for the stem marker CD133, was expanded as tumor spheroids, dissociated, and exposed in vitro to varying amounts (range 100–500 MOI) of Fn. We found that Fn stably adheres to CSCs, likely by multiple interactions involving the tumor-associated Gal-GalNac disaccharide and the Fn-docking protein CEA-family cell adhesion molecule 1 (CEACAM-1), robustly expressed on CSCs. Importantly, Fn elicited innate immune responses in CSCs and triggered a growth factor-like, protein tyrosine phosphorylation cascade largely dependent on CEACAM-1 and culminating in the activation of p42/44 MAP kinase. Thus, the direct stimulation of CSCs by Fn may contribute to microbiota-driven colorectal carcinogenesis and represent a target for innovative therapies.
Collapse
|