1
|
Altiparmak-Ulbegi G, Hasbal-Celikok G, Aksoy-Sagirli P. AKT1 and CTNNB1 mutations as drivers of paclitaxel resistance in breast cancer cells. Oncol Lett 2025; 30:324. [PMID: 40370645 PMCID: PMC12076040 DOI: 10.3892/ol.2025.15070] [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: 12/19/2024] [Accepted: 03/26/2025] [Indexed: 05/16/2025] Open
Abstract
Breast cancer (BC) is the most prevalent cancer type in the world, with increasing incidence rates. Drug resistance is a notable factor that limits the effectiveness of BC therapy. Paclitaxel (PTX), a chemotherapeutic agent belonging to the taxane class, is commonly used in BC; however, its efficacy is often compromised by drug resistance, which is primarily attributed to genetic alterations. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and wingless-type MMTV integration site family/β-catenin signaling pathways are involved in essential cellular processes, such as proliferation, apoptosis and maintenance of homeostasis. Dysregulated activation of these pathways is strongly associated with carcinogenesis and drug resistance. In the present study, the potential effects of AKT1 (E17K/E49K/L52R) and catenin β-1 (CTNNB1; S33P/T41A/S45F) mutations on PTX resistance in BC were investigated in vitro using site-directed mutagenesis, transient transfection, MTS assay and western blot analyses. The results of the present study indicated that AKT1-E17K/E49K and CTNNB1-S45F/T41A mutations induced PTX resistance compared with AKT1-wild-type (WT) and CTNNB1-WT in MCF-7 cells, respectively. In MDA-MB-231 cells, all three AKT1 mutations (E17K/E49K/L52R) triggered PTX resistance compared with AKT1-WT, while none of the CTNNB1 mutations exhibited such an effect. In conclusion, AKT1 mutations may serve as a biomarker for PTX resistance in both estrogen receptor (ER)(+)/progesterone receptor (PR)(+)/HER2(-) and triple negative BC, while CTNNB1 mutations may be a potential biomarker for PTX resistance in ER(+)/PR(+)/HER2(-) BC.
Collapse
Affiliation(s)
| | - Gozde Hasbal-Celikok
- Department of Biochemistry, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye
| | - Pinar Aksoy-Sagirli
- Department of Biochemistry, Faculty of Pharmacy, Istanbul University, 34116 Istanbul, Türkiye
| |
Collapse
|
2
|
Liu G, Qiu Y, You N, Yu M, Chen W, Sun T, Qin Z, Han M, Xue Z, Liang X, Mao B, Ling L, Wu Y, Xing W, Liu Q, Wang D. Pre-ischaemic empagliflozin treatment attenuates blood-brain barrier disruption via β-catenin mediated protection of cerebral endothelial cells. Cardiovasc Res 2025; 121:788-802. [PMID: 40173314 DOI: 10.1093/cvr/cvaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 04/04/2025] Open
Abstract
AIMS Microvascular endothelial cells dysfunction can significantly worsen ischaemic stroke outcomes by disrupting tight junctions and increasing the acquisition of adhesion molecules, accelerating blood-brain barrier (BBB) disruption and pro-inflammatory response. The identification of drugs that improve endothelial cell function may be crucial for ischaemic stroke. It has been validated that empagliflozin (EMPA), a novel antidiabetic drug, protects endothelial cells regardless of the diabetic status of the patient. However, the impact of EMPA on stroke outcomes is unclear. We hypothesized that EMPA would exert a beneficial effect on ischaemic stroke outcome by protecting microvascular endothelial cells against tight junction disruption and the increase of adhesion molecules. METHODS AND RESULTS Young adult male mice were administered with EMPA or vehicle (dimethyl sulfoxide) daily for 7 days before being subjected to transient middle cerebral artery occlusion (tMCAO). Neurological deficits were evaluated for up to 28 days post-tMCAO. Infarct volume, BBB disruption, and inflammatory status were assessed 1 day after tMCAO.bEnd.3 cells and primary brain microvascular endothelial cells were treated with EMPA or vehicle under oxygen and glucose deprivation/reperfusion (OGD/R), and the lactate dehydrogenase release, transendothelial electrical resistance, leakage of fluorescein isothiocyanate-dextran, and tight junction and adhesion molecules proteins were examined. Mechanistic studies probing the effect of EMPA on endothelial cells were conducted by RNA-seq. EMPA treatment before ischaemia markedly improved infarct volume, BBB disruption, and inflammation 1-day post-tMCAO, and further enhanced neurobehavioral function up to 28 days. Pre-treatment of EMPA attenuated endothelial cell dysfunction under OGD/R conditions. In mechanistic terms, RNA-seq data from isolated cerebral microvessels revealed that the Wnt/β-catenin signalling pathway was preserved in the EMPA group, in contrast to the vehicle group. Pre-treatment with EMPA inhibited β-catenin ubiquitination and promoted β-catenin translocation from the cytoplasm to the nucleus to improve endothelial cell function. Importantly, the β-catenin inhibitor XAV-939 eliminated this protective function of EMPA. CONCLUSION EMPA administration before tMCAO attenuated ischaemia/reperfusion-induced BBB disruption and inflammation via β-catenin-mediated protection of cerebral microvascular endothelial cells. Therefore, EMPA shows potential for improving stroke outcomes as an adjunctive preventive strategy.
Collapse
Affiliation(s)
- Guohao Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Nanlin You
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengchen Yu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenbo Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhen Qin
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhiwei Xue
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangjun Liang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Bo Mao
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lu Ling
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yanzhao Wu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenchen Xing
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Quanmeng Liu
- Department of Surgery, Shandong Provincial Hospital, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
- Department of Neurosurgery, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong 2530000, China
| |
Collapse
|
3
|
Parrish ML, Osborne-Frazier ML, Broaddus RR, Gladden AB. Differential Localization of β-Catenin Protein in CTNNB1 Mutant Endometrial Cancers Results in Distinct Transcriptional Profiles. Mod Pathol 2025; 38:100791. [PMID: 40348058 DOI: 10.1016/j.modpat.2025.100791] [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: 02/11/2025] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 05/14/2025]
Abstract
CTNNB1 exon 3 mutation is a well-established driver of nearly 30% of endometrioid endometrial cancers (EECs), and this is associated with worse patient survival. Paradoxically, we have previously demonstrated that mutant β-catenin protein does not robustly localize to the nucleus in these cancers. The purpose of this study was to determine downstream gene expression in these cancers with nuclear or membrane/cytoplasmic mutant β-catenin protein localization. Spatial transcriptomics was performed on tumors with intratumor nuclear and nonnuclear mutant β-catenin, using the protein localization to select for regions of interest (ROIs). Differential expression analysis of all nuclear and nonnuclear ROIs yielded distinct transcriptional profiles based on the localization of β-catenin. Analysis revealed enrichment for Wnt signaling and epithelial-to-mesenchymal transition pathways in nuclear ROIs and hormone signaling in nonnuclear ROIs. Hierarchical clustering yielded 2 clusters comprised of almost entirely nuclear or nonnuclear ROIs. A novel therapeutic target, TROP2, encoded by the TACSTD2 gene, was identified to be altered by Wnt/β-catenin signaling. These data provide evidence for highly heterogeneous intratumor transcriptional profiles dependent on β-catenin protein localization in EEC with CTNNB1 driver mutations. Therefore, reporting of β-catenin immunohistochemistry should include an estimated percentage of tumor with nuclear localization in EEC tumors with exon 3 CTNNB1 mutations.
Collapse
Affiliation(s)
- Molly L Parrish
- Department of Pathology and Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina; Pathobiology and Translational Science Graduate Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Macy L Osborne-Frazier
- Department of Pathology and Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina; Pathobiology and Translational Science Graduate Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Russell R Broaddus
- Department of Pathology and Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Andrew B Gladden
- Department of Pathology and Laboratory Medicine, The University of North Carolina School of Medicine, Chapel Hill, North Carolina.
| |
Collapse
|
4
|
Aguayo F, Tapia JC, Calaf GM, Muñoz JP, Osorio JC, Guzmán-Venegas M, Moreno-León C, Levican J, Andrade-Madrigal C. The Role of Xenobiotics and Anelloviruses in Colorectal Cancer: Mechanisms and Perspectives. Int J Mol Sci 2025; 26:4354. [PMID: 40362591 PMCID: PMC12072659 DOI: 10.3390/ijms26094354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025] Open
Abstract
Xenobiotics are non-natural chemical compounds to which the human population is exposed. Chronic exposure to certain xenobiotics is associated with various diseases, including cancer development. Anelloviruses (AVs), including Torque Teno Virus (TTV), Torque Teno Mini Virus (TTMV), and Torque Teno Midi Virus (TTMDV), are ubiquitous viruses found in the general population. As no disease has been definitively associated with AVs, they are sometimes referred to as "viruses awaiting a disease". This review explores the potential roles of xenobiotics and AVs in colorectal cancer (CRC) development and suggests a potential interplay between them. Evidence suggests an association between certain xenobiotics (like pesticides, cigarette smoke components, and dietary factors) and CRC, while such an association is less clear for AVs. The high prevalence of AVs suggests these infections alone may be insufficient to disrupt homeostasis; thus, additional factors might be required to promote disease, potentially including cancer.
Collapse
Affiliation(s)
- Francisco Aguayo
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Julio C. Tapia
- Laboratorio de Transformación Celular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Santiago 8380453, Chile
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile;
| | - Julio C. Osorio
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Matías Guzmán-Venegas
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Carolina Moreno-León
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| | - Jorge Levican
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Cristian Andrade-Madrigal
- Laboratorio de Oncovirología, Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad de Tarapacá, Arica 1000000, Chile; (J.C.O.); (M.G.-V.); (C.M.-L.); (C.A.-M.)
| |
Collapse
|
5
|
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
|
6
|
Li L, Kang Y, Cheng R, Liu F, Wu F, Liu Z, Kou J, Zhang Z, Li W, Zhao H, He X, Du W. The de novo synthesis of GABA and its gene regulatory function control hepatocellular carcinoma metastasis. Dev Cell 2025; 60:1053-1069.e6. [PMID: 39740661 DOI: 10.1016/j.devcel.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/14/2024] [Accepted: 12/04/2024] [Indexed: 01/02/2025]
Abstract
The neurotransmitter gamma-aminobutyric acid (GABA) has been thought to be involved in the development of some types of cancer. Yet, the de novo synthesis of GABA and how it functions in hepatocellular carcinoma (HCC) remain unclear. Here, we report that SLC6A12 acts as a transporter of GABA, and that aldehyde dehydrogenase 9 family member A1 (ALDH9A1), not glutamate decarboxylase 1 (GAD1), generates GABA in human HCC. Interestingly, SLC6A12 and ALDH9A1 are upregulated during lung metastases of HCC, and depletion of either of them leads to impaired HCC metastasis. Mechanistically, GABA directly binds and stabilizes β-catenin, resulting in activated Wnt/β-catenin signaling, and thereby enhancing HCC metastasis. Reciprocally, β-catenin transcriptionally upregulates SLC6A12 to import more GABA to stabilize β-catenin. Thus, our findings identify ALDH9A1 as the major GABA synthetase in HCC, demonstrate a positive-feedback regulatory mechanism for sustaining Wnt/β-catenin signaling, and reveal a role for β-catenin in sensing GABA, which contributes to HCC metastasis.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Youli Kang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Running Cheng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Fangming Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Fujia Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zizhao Liu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Junjie Kou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Zhenxi Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Wei Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Haitao Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing 100730, China.
| | - Xiaojing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases, Haihe Laboratory of Cell Ecosystem, Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
7
|
Terra ML, Sant’Anna TBF, de Barros JJF, de Araujo NM. Geographic and Viral Etiology Patterns of TERT Promoter and CTNNB1 Exon 3 Mutations in Hepatocellular Carcinoma: A Comprehensive Review. Int J Mol Sci 2025; 26:2889. [PMID: 40243493 PMCID: PMC11988703 DOI: 10.3390/ijms26072889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and a leading cause of cancer-related mortality worldwide. Genetic alterations play a critical role in hepatocarcinogenesis, with mutations in the telomerase reverse transcriptase promoter (TERTp) and CTNNB1 exon 3 representing two of the most frequently reported somatic events in HCC. However, the frequency and distribution of these mutations vary across geographic regions and viral etiologies, particularly hepatitis B virus (HBV) and hepatitis C virus (HCV). This study aimed to assess the global distribution and etiological associations of TERTp and CTNNB1 exon 3 mutations in HCC through a comprehensive literature review. Our analysis, encompassing over 4000 HCC cases, revealed that TERTp mutations were present in 49.2% of tumors, with C228T being the predominant variant (93.3% among mutated cases). A striking contrast was observed between viral etiologies: TERTp mutations were detected in 31.6% of HBV-related HCCs, compared to 66.2% in HCV-related cases. CTNNB1 exon 3 mutations were identified in 23.1% of HCCs, showing a similar association with viral etiology, being more common in HCV-related cases (30.7%) than in HBV-related tumors (12.8%). Geographically, both mutations exhibited comparable patterns, with higher frequencies in Europe, Japan, and the USA, while lower rates were observed in China, Taiwan, and South Korea. Our findings underscore the distinct molecular profiles of HCC according to viral etiology and geographic origin, highlighting the need for region- and etiology-specific approaches to HCC prevention, diagnosis, and targeted therapy.
Collapse
Affiliation(s)
| | | | | | - Natalia Motta de Araujo
- Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil; (M.L.T.); (T.B.F.S.); (J.J.F.d.B.)
| |
Collapse
|
8
|
Vasu M, Ahlawat S, Arora R, Sharma R. Deciphering the molecular drivers for cashmere/pashmina fiber production in goats: a comprehensive review. Mamm Genome 2025; 36:162-182. [PMID: 39904908 DOI: 10.1007/s00335-025-10109-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
Cashmere, also known as pashmina, is derived from the secondary hair follicles of Cashmere/Changthangi goats. Renowned as the world's most luxurious natural fiber, it holds significant economic value in the textile industry. This comprehensive review enhances our understanding of the complex biological processes governing cashmere/pashmina fiber development and quality, enabling advancements in selective breeding and fiber enhancement strategies. The review specifically examines the molecular determinants influencing fiber development, with an emphasis on keratins (KRTs) and keratin-associated proteins (KRTAPs). It also explores the roles of key molecular pathways, including Wnt, Notch, BMP, NF-kappa B, VEGF, cAMP, PI3K-Akt, ECM, cell adhesion, Hedgehog, MAPK, Ras, JAK-STAT, TGF-β, mTOR, melanogenesis, FoxO, Hippo, and Rap1 signaling. Understanding these intricate molecular cascades provides valuable insights into the mechanisms orchestrating hair follicle growth, further advancing the biology of this coveted natural fiber. Expanding multi-omics approaches will enhance breeding precision and deepen our understanding of molecular pathways influencing cashmere production. Future research should address critical gaps, such as the impact of environmental factors, epigenetic modifications, and functional studies of genetic variants. Collaboration among breeders, researchers, and policymakers is essential for translating genomic advancements into practical applications. Such efforts can promote sustainable practices, conserve biodiversity, and ensure the long-term viability of high-quality cashmere production. Aligning genetic insights with conservation strategies will support the sustainable growth of the cashmere industry while preserving its economic and ecological value.
Collapse
Affiliation(s)
- Mahanthi Vasu
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Sonika Ahlawat
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India.
| | - Reena Arora
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| | - Rekha Sharma
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, India
| |
Collapse
|
9
|
Nam KY, Kim MS, An J, Min S, Lee JH, Park JS, Huh C, Yun SH, Lee KJ. Human-Centric, Three Dimensional Micro Light-Emitting Diodes for Cosmetic and Medical Phototherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416716. [PMID: 39960366 PMCID: PMC11905057 DOI: 10.1002/advs.202416716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/03/2025] [Indexed: 03/14/2025]
Abstract
Phototherapy based on micro light-emitting diodes (µLEDs) has gained enormous attention in the medical field as a patient-friendly therapeutic method due to its advantages of minimal invasiveness, fewer side effects, and versatile device form factors with high stability in biological environment. Effective cosmetic and medical phototherapy depends on deep light penetration, precise irradiation, and simultaneous multi-site stimulation, facilitated by three-dimensional (3D) optoelectronics specifically designed for complex human matters, defined here as 3D µLEDs. This perspective article aims to present the functionalities and strategies of 3D µLEDs for human-centric phototherapy. This study investigates the effectiveness of phototherapy enabled by three key functionalities such as shape morphing, self-adaptation, and multilayered spatiotemporal mapping of 3D µLEDs. Finally, this article provides future insights of 3D µLEDs for human-centric phototherapy applications.
Collapse
Affiliation(s)
- Ki Yun Nam
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
- School of Electrical EngineeringGraduate School of Semiconductor TechnologyKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
| | - Min Seo Kim
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
| | - Jaehun An
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
| | - Seongwook Min
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
| | - Jae Hee Lee
- Querrey‐Simpson Institute for BioelectronicsNorthwestern UniversityEvanstonIL60 208USA
| | - Jae Sung Park
- Yonsei Myview Clinic301, Sadang‐ro, Dongjak‐guSeoul0 7008Republic of Korea
| | - Chang‐Hun Huh
- Department of DermatologySeoul National University Bundang Hospital (SNUBH)173–82, Gumi‐ro, Bundang‐guSeongnam13 620Republic of Korea
| | - Seok Hyun Yun
- Harvard Medical School and Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA0 2114USA
| | - Keon Jae Lee
- Department of Materials Science and EngineeringKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
- School of Electrical EngineeringGraduate School of Semiconductor TechnologyKorea Advanced Institute of Science and Technology (KAIST)291 Daehak‐ro, Yuseong‐guDaejeon34 141Republic of Korea
| |
Collapse
|
10
|
Yao CY, Tao HT, He JJ, Zhu FY, Xie CQ, Cheng YN, Li JQ, Liu ZZ, Hou CY, Liu XL, Fan YL, Fang D, Lv XR. NUAK1 acts as a novel regulator of PD-L1 via activating GSK-3β/β-catenin pathway in hepatocellular carcinoma. Mol Med 2025; 31:38. [PMID: 39901136 PMCID: PMC11789290 DOI: 10.1186/s10020-025-01088-7] [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: 05/30/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND NUAK1 is associated with metastasis and drug resistance in hepatocellular carcinoma (HCC). However, little is known about the immune functions of NUAK1 in HCC. Therefore, the aim of this study was to elucidate the novel role of NUAK1 in facilitating immune evasion in HCC and to investigate the mechanisms underpinning this process. METHOD The levels of NUAK1 expression and the infiltration of CD8+ T cells were assessed in tumor tissues from HCC patients and mice xenograft model. HCC cell lines were used to validate the role of NUAK1 in regulating the transcription of PD-L1, the diethylnitrosamine-induced HCC model was established and the expression levels of NUAK1 and PD-L1 proteins in the rat livers were detected. Western blotting, immunofluorescence, real time PCR, and immunohistochemical staining were used to investigate the underlying mechanisms by which NUAK1 regulates PD-L1 expression in hepatocellular carcinoma. RESULTS NUAK1 expression was negatively correlated with CD8+ T cell infiltration in tumor tissues from HCC patients and mice xenograft model. Both gain and loss of functions have identified NUAK1 promoted PD-L1 expression at transcriptional level in HCC cells. The increased expression of NUAK1 and PD-L1 proteins were observed in the rat livers of diethylnitrosamine-induced HCC model. Moreover, overexpression of NUAK1 promotes GSK3β Ser9 phosphorylation, β-catenin expression and nuclear accumulation in HCC cells. By contrast, knockdown of NUAK1 has opposite effects. Inhibition of GSK3β activity significantly promoted β-catenin expression and PD-L1 expression in HCC cells. IHC analyses of tumor tissues from HCC patients suggested that the levels of p-GSK3β and β-catenin were positively correlated with NUAK1 expression. Knockdown of β-catenin also reversed NUAK1-mediated PD-L1 expression in HCC cells. CONCLUSIONS This study revealed a novel role for NUAK1, which promotes the transcriptional expression of PD-L1 by activating GSK3β/β-catenin signaling pathway, leading to immune escape of hepatocellular carcinoma. Registry and the registration no. of the study/trial: Not applicable.
Collapse
Affiliation(s)
- Chao-Yan Yao
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Hang-Tian Tao
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Jin-Jin He
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
| | - Feng-Yi Zhu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Cui-Qing Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yu-Na Cheng
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Ji-Qin Li
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zhuang-Zhuang Liu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Chun-Yu Hou
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Xue-Li Liu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yong-Li Fan
- Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475000, China.
| | - Dong Fang
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China.
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China.
| | - Xin-Rui Lv
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, The First Affiliated Hospital of Henan University, Ximen Ave, Kaifeng, 475000, China.
| |
Collapse
|
11
|
Warrier VP, Venkatachalam S, Sakthivel R, Gromiha MM, Karunagaran D. Combinatorial Effects of 5-Fluorouracil and Menadione on Wnt/β-Catenin Pathway in Human Colorectal Cancer Cells. Appl Biochem Biotechnol 2025; 197:1280-1300. [PMID: 39404999 DOI: 10.1007/s12010-024-05072-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 02/13/2025]
Abstract
The incidence and mortality rates of colorectal cancer (CRC) are alarmingly high, and the scientific community is consistently engaged in developing newer therapeutic options for cancer cure or prevention. The fluoropyrimidine drug, 5-fluorouracil (5FU), remains the first line of treatment against CRC; nevertheless, relapses frequently occur since the cells gain resistance over time through various mechanisms. Studies have highlighted the significance of combinatorial treatment of a Wnt signaling inhibitor and 5FU as a better treatment strategy to overcome 5FU resistance. Small molecules that specifically target and disrupt β-catenin-TCF interaction, a crucial step of the Wnt signaling, are promising in CRC treatment. In this study, we investigated the synergistic cytotoxic activity of menadione with 5FU as the former has previously been shown to downregulate Wnt signaling in CRC cells. Docking and experimental results suggest that the drug combination interfered with key protein-protein interactions in the β-catenin-TCF complex, exerted synergistic anti-cancerous effects in CRC cells, and downregulated the expression of Wnt signaling proteins. Taken together, our data suggest that the simultaneous binding of 5FU and menadione to β-catenin can block Wnt signaling by disrupting β-catenin-TCF interaction and inhibit the proliferation of CRC cells.
Collapse
Affiliation(s)
- Vidya P Warrier
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Sankaran Venkatachalam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Ramasamy Sakthivel
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| |
Collapse
|
12
|
Tsai YW, Tseng YS, Wu YS, Song WL, You MY, Hsu YC, Chen WP, Huang WH, Chng JC, Lim CL, Wei KH, Ben Lai SL, Lee WC, Yang KC. N-Cadherin promotes cardiac regeneration by potentiating pro-mitotic β-Catenin signaling in cardiomyocytes. Nat Commun 2025; 16:896. [PMID: 39837836 PMCID: PMC11751462 DOI: 10.1038/s41467-025-56216-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
Adult human hearts exhibit limited regenerative capacity. Post-injury cardiomyocyte (CM) loss can lead to myocardial dysfunction and failure. Although neonatal mammalian hearts can regenerate, the underlying molecular mechanisms remain elusive. Herein, comparative transcriptome analyses identify adherens junction protein N-Cadherin as a crucial regulator of CM proliferation/renewal. Its expression correlates positively with mitotic genes and shows an age-dependent reduction. N-Cadherin is upregulated in the neonatal mouse heart following injury, coinciding with increased CM mitotic activities. N-Cadherin knockdown reduces, whereas overexpression increases, the proliferation activity of neonatal mouse CMs and human induced pluripotent stem cell-derived CMs. Mechanistically, N-Cadherin binds and stabilizes pro-mitotic transcription regulator β-Catenin, driving CM self-renewal. Targeted N-Cadherin deletion in CMs impedes cardiac regeneration in neonatal mice, leading to excessive scarring. N-Cadherin overexpression, by contrast, promotes regeneration in adult mouse hearts following ischemic injury. N-Cadherin targeting presents a promising avenue for promoting cardiac regeneration and restoring function in injured adult human hearts.
Collapse
Grants
- This work was funded by Taiwan National Science and Technology Council Grants 111-2628-B-002-008, 111-2314-B-002-069 MY3, 112-2314-B-002-277 MY3, 112-2918-I-002-002 and 112-2926-I-002-511-G (KCY), an Innovative Research Grant from Taiwan National Health Research Institute NHRI-EX112-11213BI (KCY), a CRC Translational Research Grant IBMS-CRC111-P01 (KCY & SLL) and a Grand Challenge Program Grant AS-GC-110-L06 (KCY & SLL) from Academia Sinica, Taiwan, grants from National Taiwan University Hospital NTUH. VN111-08, VN112-06, VN-113-03, 111-S0042, 112-S0307, 112-S0311, 113-S0196, 111-IF0005, 113-IF0002, 113-E0008 (KCY), Collaborative Research Projects of National Taiwan University College of Medicine, National Taiwan University Hospital and Min-Sheng General Hospital 109F005-110-B3, 109F005-111-C2, 119F005-112-M2 (KCY), grants from the Excellent Translation Medicine Research Projects of National Taiwan University College of Medicine and National Taiwan University Hospital, NSCCMOH-131-41, 111C101-051, 112C101-031 (KCY) and Career Development Grants from National Taiwan University 112L7849, 113L7832 (KCY).
Collapse
Affiliation(s)
- Yi-Wei Tsai
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Yi-Shuan Tseng
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Yu-Shuo Wu
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Wei-Lun Song
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Min-Yi You
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Yun-Chia Hsu
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Wen-Pin Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Wei-Han Huang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
| | - Jia-Ci Chng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Chai-Ling Lim
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Ke-Hsuan Wei
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shih-Lei Ben Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Wen-Chih Lee
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan, ROC.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC.
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC.
- Research Center for Developmental Biology & Regenerative Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Center for Frontier Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| |
Collapse
|
13
|
Lainšček D, Forstnerič V, Miroševič Š. CTNNB1 syndrome mouse models. Mamm Genome 2025:10.1007/s00335-025-10105-3. [PMID: 39833474 DOI: 10.1007/s00335-025-10105-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
CTNNB1 syndrome is a rare neurodevelopmental disorder, affecting children worldwide with a prevalence of 2.6-3.2 per 100,000 births and often misdiagnosed as cerebral palsy. De novo loss-of-function mutations in the Ctnnb1 gene result in dysfunction of the β-catenin protein, disrupting the canonical Wnt signaling pathway, which plays a key role in cell proliferation, differentiation, and tissue homeostasis. Additionally, these mutations impair the formation of cell junctions, adversely affecting tissue architecture. Motor and speech deficits, cognitive impairment, cardiovascular and visual problems are just some of the key symptoms that occur in CTNNB1 syndrome patients. There is currently no effective treatment option available for patients with CTNNB1 syndrome, with support largely focused on the management of symptoms and physiotherapy, yet recently some therapeutic approaches are being developed. Animal testing is still crucial in the process of new drug development, and mouse models are particularly important. These models provide researchers with new understanding of the disease mechanisms and are invaluable for testing the efficacy and safety of potential treatments. The development of various mouse models with β-catenin loss- and gain-of-function mutations successfully replicates key features of intellectual disability, autism-like behaviors, motor deficits, and more. These models provide a valuable platform for studying disease mechanisms and offer a powerful tool for testing the therapeutic potential and effectiveness of new drug candidates, paving the way for future clinical trials.
Collapse
Affiliation(s)
- Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, 1000, Slovenia.
- Centre for Technologies of Gene and Cell Therapy, Ljubljana, 1000, Slovenia.
- EN-FIST Centre of Excellence, Ljubljana, 1000, Slovenia.
| | - Vida Forstnerič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Ljubljana, 1000, Slovenia.
| | - Špela Miroševič
- The Gene Therapy Research Institute, CTNNB1 Foundation, Ljubljana, 1000, Slovenia.
- Department of Family Medicine, Faculty of Medicine Ljubljana, University of Ljubljana, Ljubljana, 1000, Slovenia.
| |
Collapse
|
14
|
Dantzer C, Dif L, Vaché J, Basbous S, Billottet C, Moreau V. Specific features of ß-catenin-mutated hepatocellular carcinomas. Br J Cancer 2024; 131:1871-1880. [PMID: 39261716 PMCID: PMC11628615 DOI: 10.1038/s41416-024-02849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
CTNNB1, encoding the ß-catenin protein, is a key oncogene contributing to liver carcinogenesis. Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer in adult, representing the third leading cause of cancer-related death. Aberrant activation of the Wnt/ß-catenin pathway, mainly due to mutations of the CTNNB1 gene, is observed in a significant subset of HCC. In this review, we first resume the major recent advances in HCC classification with a focus on CTNNB1-mutated HCC subclass. We present the regulatory mechanisms involved in β-catenin stabilisation, transcriptional activity and binding to partner proteins. We then describe specific phenotypic characteristics of CTNNB1-mutated HCC thanks to their unique gene expression patterns. CTNNB1-mutated HCC constitute a full-fledged subclass of HCC with distinct pathological features such as well-differentiated cells with low proliferation rate, association to cholestasis, metabolic alterations, immune exclusion and invasion. Finally, we discuss therapeutic approaches to target ß-catenin-mutated liver tumours and innovative perspectives for future drug developments.
Collapse
Affiliation(s)
| | - Lydia Dif
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | - Justine Vaché
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | - Sara Basbous
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | | | | |
Collapse
|
15
|
Wang X, Qu Z, Zhao S, Luo L, Yan L. Wnt/β-catenin signaling pathway: proteins' roles in osteoporosis and cancer diseases and the regulatory effects of natural compounds on osteoporosis. Mol Med 2024; 30:193. [PMID: 39468464 PMCID: PMC11520425 DOI: 10.1186/s10020-024-00957-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Osteoblasts are mainly derived from mesenchymal stem cells in the bone marrow. These stem cells can differentiate into osteoblasts, which have the functions of secreting bone matrix, promoting bone formation, and participating in bone remodeling. The abnormality of osteoblasts can cause a variety of bone-related diseases, including osteoporosis, delayed fracture healing, and skeletal deformities. In recent years, with the side effects caused by the application of PTH drugs, biphosphonate drugs, and calmodulin drugs, people have carried out more in-depth research on the mechanism of osteoblast differentiation, and are actively looking for natural compounds for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway is considered to be one of the important pathways of osteoblast differentiation, and has become an important target for the treatment of osteoporosis. The Wnt/β-catenin signaling pathway, whether its activation is enhanced or its expression is weakened, will cause a variety of diseases including tumors. This review will summarize the effect of Wnt/β-catenin signaling pathway on osteoblast differentiation and the correlation between the related proteins in the pathway and human diseases. At the same time, the latest research progress of natural compounds targeting Wnt/β-catenin signaling pathway against osteoporosis is summarized.
Collapse
Affiliation(s)
- Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
- Xi'an Medical University, Xi'an, China
| | - Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lei Luo
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
16
|
Wu Y, Yang J, Wang X, Guo J, Tan Z, Guan F, Cao L. NCAM and attached polysialic acid affect behaviors of breast epithelial cells through differential signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39420834 DOI: 10.3724/abbs.2024176] [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] [Indexed: 10/19/2024] Open
Abstract
Neural cell adhesion molecule (NCAM), a common mammalian cell surface glycoprotein, is the major substrate of polysialic acid (polySia). Polysialylated NCAM occurs in many types of cancer, but rarely in normal adult tissues. The functional role of NCAM hypersialylation in the epithelial-mesenchymal transition (EMT) process remains unclear. The present study indicates that NCAM and attached polysialic acid affect behaviors of breast epithelial cells through differential signaling pathways. NCAM and polysialylated NCAM are aberrantly regulated in breast cancer cells. They are both upregulated in normal breast epithelial cells undergoing EMT. Western blot analysis demonstrates that NCAM-140 overexpression induces EMT in breast epithelial cells and promotes cell proliferation and migration through activation of the β-catenin/slug signaling pathway. Modification of polySia attached to NCAM modulates cell adhesion and promotes cell motility through activation of the EGFR/STAT3 pathway. These observations contribute to clarifying the molecular mechanisms by which polysialic acid and its major substrate, NCAM, modulate cell behaviors, and highlight the significance of increased polysialylated expression on NCAM during EMT and tumor development.
Collapse
Affiliation(s)
- Yurong Wu
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Juhong Yang
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xin Wang
- Shandong University of Arts, Ji'nan 250300, China
| | - Jia Guo
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China
| | - Zengqi Tan
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, School of Medicine, Northwest University, Xi'an 710069, China
| | - Feng Guan
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Lin Cao
- Provincial Key Laboratory of Biotechnology, Joint International Research Laboratory of Glycobiology and Medicinal Chemistry, School of Medicine, Northwest University, Xi'an 710069, China
| |
Collapse
|
17
|
Samant C, Kale R, Pai KSR, Nandakumar K, Bhonde M. Role of Wnt/β-catenin pathway in cancer drug resistance: Insights into molecular aspects of major solid tumors. Biochem Biophys Res Commun 2024; 729:150348. [PMID: 38986260 DOI: 10.1016/j.bbrc.2024.150348] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Adaptive resistance to conventional and targeted therapies remains one of the major obstacles in the effective management of cancer. Aberrant activation of key signaling mechanisms plays a pivotal role in modulating resistance to drugs. An evolutionarily conserved Wnt/β-catenin pathway is one of the signaling cascades which regulate resistance to drugs. Elevated Wnt signaling confers resistance to anticancer therapies, either through direct activation of its target genes or via indirect mechanisms and crosstalk over other signaling pathways. Involvement of the Wnt/β-catenin pathway in cancer hallmarks like inhibition of apoptosis, promotion of invasion and metastasis and cancer stem cell maintenance makes this pathway a potential target to exploit for addressing drug resistance. Accumulating evidences suggest a critical role of Wnt/β-catenin pathway in imparting resistance across multiple cancers including PDAC, NSCLC, TNBC, etc. Here we present a comprehensive assessment of how Wnt/β-catenin pathway mediates cancer drug resistance in majority of the solid tumors. We take a deep dive into the Wnt/β-catenin signaling-mediated modulation of cellular and downstream molecular mechanisms and their impact on cancer resistance.
Collapse
Affiliation(s)
- Charudatt Samant
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India.
| | - Ramesh Kale
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Mandar Bhonde
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| |
Collapse
|
18
|
Enos MD, Gavagan M, Jameson N, Zalatan JG, Weis WI. Structural and functional effects of phosphopriming and scaffolding in the kinase GSK-3β. Sci Signal 2024; 17:eado0881. [PMID: 39226374 PMCID: PMC11461088 DOI: 10.1126/scisignal.ado0881] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Glycogen synthase kinase 3β (GSK-3β) targets specific signaling pathways in response to distinct upstream signals. We used structural and functional studies to dissect how an upstream phosphorylation step primes the Wnt signaling component β-catenin for phosphorylation by GSK-3β and how scaffolding interactions contribute to this reaction. Our crystal structure of GSK-3β bound to a phosphoprimed β-catenin peptide confirmed the expected binding mode of the phosphoprimed residue adjacent to the catalytic site. An aspartate phosphomimic in the priming site of β-catenin adopted an indistinguishable structure but reacted approximately 1000-fold slower than the native phosphoprimed substrate. This result suggests that substrate positioning alone is not sufficient for catalysis and that native phosphopriming interactions are necessary. We also obtained a structure of GSK-3β with an extended peptide from the scaffold protein Axin that bound with greater affinity than that of previously crystallized Axin fragments. This structure neither revealed additional contacts that produce the higher affinity nor explained how substrate interactions in the GSK-3β active site are modulated by remote Axin binding. Together, our findings suggest that phosphopriming and scaffolding produce small conformational changes or allosteric effects, not captured in the crystal structures, that activate GSK-3β and facilitate β-catenin phosphorylation. These results highlight limitations in our ability to predict catalytic activity from structure and have potential implications for the role of natural phosphomimic mutations in kinase regulation and phosphosite evolution.
Collapse
Affiliation(s)
- Michael D. Enos
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Maire Gavagan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Noel Jameson
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jesse G. Zalatan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - William I. Weis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| |
Collapse
|
19
|
Stewart RA, Ding Z, Jeon US, Goodman LB, Tran JJ, Zientko JP, Sabu M, Cadigan KM. Wnt target gene activation requires β-catenin separation into biomolecular condensates. PLoS Biol 2024; 22:e3002368. [PMID: 39316611 PMCID: PMC11460698 DOI: 10.1371/journal.pbio.3002368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/08/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
The Wnt/β-catenin signaling pathway plays numerous essential roles in animal development and tissue/stem cell maintenance. The activation of genes regulated by Wnt/β-catenin signaling requires the nuclear accumulation of β-catenin, a transcriptional co-activator. β-catenin is recruited to many Wnt-regulated enhancers through direct binding to T-cell factor/lymphoid enhancer factor (TCF/LEF) family transcription factors. β-catenin has previously been reported to form phase-separated biomolecular condensates (BMCs), which was implicated as a component of β-catenin's mechanism of action. This function required aromatic amino acid residues in the intrinsically disordered regions (IDRs) at the N- and C-termini of the protein. In this report, we further explore a role for β-catenin BMCs in Wnt target gene regulation. We find that β-catenin BMCs are miscible with LEF1 BMCs in vitro and in cultured cells. We characterized a panel of β-catenin mutants with different combinations of aromatic residue mutations in human cell culture and Drosophila melanogaster. Our data support a model in which aromatic residues across both IDRs contribute to BMC formation and signaling activity. Although different Wnt targets have different sensitivities to loss of β-catenin's aromatic residues, the activation of every target examined was compromised by aromatic substitution. These mutants are not defective in nuclear import or co-immunoprecipitation with several β-catenin binding partners. In addition, residues in the N-terminal IDR with no previously known role in signaling are clearly required for the activation of various Wnt readouts. Consistent with this, deletion of the N-terminal IDR results in a loss of signaling activity, which can be rescued by the addition of heterologous IDRs enriched in aromatic residues. Overall, our work supports a model in which the ability of β-catenin to form biomolecular condensates in the nucleus is tightly linked to its function as a transcriptional co-regulator.
Collapse
Affiliation(s)
- Richard A. Stewart
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zhihao Ding
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ung Seop Jeon
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lauren B. Goodman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jeannine J. Tran
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John P. Zientko
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Malavika Sabu
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ken M. Cadigan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
20
|
Sun M, Lu Y, Zhang H, Jiang W, Wang W, Huang X, Zhang S, Xiang D, Tang B, Chen Y, Chen T, Lian C, Zhang J. Multifunctional Composite Scaffold with Nanosilver, Graphene Oxide, and Macrophage Membrane Vesicles for Sequential Treatment of Infected Bone Defects. Adv Healthc Mater 2024; 13:e2400346. [PMID: 38684106 DOI: 10.1002/adhm.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/02/2024] [Indexed: 05/02/2024]
Abstract
The management of infected bone defects poses a significant clinical challenge, and current treatment modalities exhibit various limitations. This study focuses on the development of a multifunctional composite scaffold comprising nanohydroxyapatite/polyethyleneglycol diacrylate matrix, silver nanoparticles, graphene oxide (GO), sodium alginate, and M2-type macrophage membrane vesicles (MVs) to enhance the healing of infected bone defects. The composite scaffold demonstrates several key features: first, it releases sufficient quantities of silver ions to effectively eliminate bacteria; second, the controlled release of MVs leads to a notable increase in M2-type macrophages, thereby significantly mitigating the inflammatory response. Additionally, GO acts synergistically with nanohydroxyapatite to enhance osteoinductive activity, thereby fostering bone regeneration. Through meticulous in vitro and in vivo investigations, the composite scaffold exhibits broad-spectrum antimicrobial effects, robust immunomodulatory capabilities, and enhanced osteoinductive activity. This multifaceted composite scaffold presents a promising approach for the sequential treatment of infected bone defects, addressing the antimicrobial, immunomodulatory, and osteogenic aspects. This study introduces innovative perspectives and offers new and effective treatment alternatives for managing infected bone defects.
Collapse
Affiliation(s)
- Mingjie Sun
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yang Lu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Hongrui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weiqian Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 401336, China
| | - Wenzhao Wang
- Department of Orthopedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xiao Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shichun Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 401336, China
| | - Dulei Xiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Boyu Tang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tingmei Chen
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jian Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
21
|
Yu L, Huang L, Zhao Y, Liu S, Zhou R, Yue Y, Sun H, Su X, Liu Q, Li S, Ying J, Zhao F, Qu Y. Atorvastatin Promotes Pro/anti-inflammatory Phenotypic Transformation of Microglia via Wnt/β-catenin Pathway in Hypoxic-Ischemic Neonatal Rats. Mol Neurobiol 2024; 61:3559-3577. [PMID: 37996729 PMCID: PMC11087325 DOI: 10.1007/s12035-023-03777-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023]
Abstract
Inflammatory reaction plays a key role in the pathogenesis of hypoxic-ischemic encephalopathy (HIE) in neonates. Microglia are resident innate immune cells in the central nervous system and are profoundly involved in neuroinflammation. Studies have revealed that atorvastatin exerts a neuroprotective effect by regulating neuroinflammation in adult animal models of brain stroke and traumatic brain injury, but its role regarding damage to the developing brain remains unclear. This study aimed to clarify the effect and mechanism of atorvastatin on the regulation of microglia function in neonatal hypoxic-ischemic brain damage (HIBD). The oxygen glucose deprivation (OGD) of microglia and neonatal rat HIBD model was established. Atorvastatin, recombinant sclerostin protein (SOST), and XAV939 (degradation of β-catenin) were administered to OGD microglia and HIBD rats. The pathological changes of brain tissue, cerebral infarction volume, learning and memory ability of rats, pro-inflammatory (CD16+/Iba1+) and anti-inflammatory (CD206+/Iba1+) microglia markers, inflammation-related indicators (Inos, Tnfα, Il6, Arg1, Tgfb, and Mrc1), and Wnt/β-catenin signaling molecules were examined. Atorvastatin reduced OGD-induced pro-inflammatory microglia and pro-inflammatory factors, while increasing anti-inflammatory microglia and anti-inflammatory factors. In vivo, atorvastatin attenuated hypoxia-ischemia (HI)-induced neuroinflammation and brain damage. Mechanistically, atorvastatin decreased SOST expression and activated the Wnt/β-catenin signaling pathway, and the administration of recombinant SOST protein or XAV939 inhibited Wnt/β-catenin signaling and attenuated the anti-inflammatory effect of atorvastatin. Atorvastatin promotes the pro/anti-inflammatory phenotypic transformation of microglia via the Wnt/β-catenin pathway in HI neonatal rats. Atorvastatin may be developed as a potent agent for the treatment of HIE in neonates.
Collapse
Affiliation(s)
- Luting Yu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lingyi Huang
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China College of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Yuanyuan Zhao
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shixi Liu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ruixi Zhou
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Yue
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Hao Sun
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaojuan Su
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qian Liu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shiping Li
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Junjie Ying
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Fengyan Zhao
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Qu
- Department of Pediatrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Ministry of Education), NHC Key Laboratory of Chronobiology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
22
|
Gottlieb S, van der Vaart A, Hassan A, Bledsoe D, Morgan A, O'Rourke B, Rogers WD, Wolstenholme JT, Miles MF. A selective GSK3β inhibitor, tideglusib, decreases intermittent access and binge ethanol self-administration in C57BL/6J mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593949. [PMID: 38798478 PMCID: PMC11118361 DOI: 10.1101/2024.05.13.593949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Over 10% of the US population over 12 years old meets criteria for Alcohol Use Disorder (AUD), yet few effective, long-term treatments are currently available. Glycogen synthase kinase 3 beta (GSK3β) has been implicated in ethanol behaviors and poses as a potential therapeutic target in the treatment of AUD. Here we investigate the role of tideglusib, a selective GSK3β inhibitor, in ethanol consumption and other behaviors. We have shown tideglusib decreases ethanol consumption in both a model of daily, progressive ethanol intake (two-bottle choice, intermittent ethanol access) and binge-like drinking behavior (drinking-in-the-dark) without effecting water intake. Further, we have shown tideglusib to have no effect on ethanol pharmacokinetics, taste preference, or anxiety-like behavior, though there was a transient increase in total locomotion following treatment. Additionally, we assessed liver health following treatment via serum levels of alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase and showed no effect on aminotransferase levels though there was a decrease in alkaline phosphatase. RNA sequencing studies revealed a role of GSK3β inhibition via tideglusib on the canonical Wnt signaling pathway, suggesting tideglusib may carry out its effects on ethanol consumption through effects on β-catenin binding to the transcription factors TCF3 and LEF1. The data presented here further implicate GSK3β in alcohol consumption and support the use of tideglusib as a potential therapeutic in the treatment of AUD.
Collapse
|
23
|
Roy S, Mehta D, Paradkar A, Chovatiya G, Waghmare SK. Dab2 (Disabled-2), an adaptor protein, regulates self-renewal of hair follicle stem cells. Commun Biol 2024; 7:525. [PMID: 38702433 PMCID: PMC11068889 DOI: 10.1038/s42003-024-06047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/13/2024] [Indexed: 05/06/2024] Open
Abstract
Disabled 2 (Dab2), an adaptor protein, is up regulated in the hair follicle stem cells (HFSCs); however, its role in any tissue stem cells has not been studied. In the present study, we have reported that Dab2 conditional knockout (Dab2-cKO) mice exhibited a delay in the HF cycle due to perturbed activation of HFSCs. Further, Dab2-cKO mice showed a reduction in the number of HFSCs and reduced colony forming ability of HFSCs. Dab2-cKO mice showed extended quiescence of HFSCs concomitant with an increased expression of Nfatc1. Dab2-cKO mice showed a decreased expression of anti-aging genes such as Col17a1, decorin, Sirt2 and Sirt7. Dab2-cKO mice did not show full hair coat recovery in aged mice thereby suggesting an accelerated aging process. Overall, we unveil for the first time, the role of Dab2 that regulate activation and self-renewal of HFSCs.
Collapse
Affiliation(s)
- Sayoni Roy
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Darshan Mehta
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Akshay Paradkar
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Gopal Chovatiya
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Sanjeev K Waghmare
- Stem Cell Biology Group, Waghmare Lab, Cancer Research Institute, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, Maharashtra, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India.
| |
Collapse
|
24
|
Lin C, Yu M, Wu X, Wang H, Wei M, Zhang L. Targeting Moonlighting Enzymes in Cancer. Molecules 2024; 29:1573. [PMID: 38611852 PMCID: PMC11013064 DOI: 10.3390/molecules29071573] [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: 03/15/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Moonlighting enzymes are multifunctional proteins that perform multiple functions beyond their primary role as catalytic enzymes. Extensive research and clinical practice have demonstrated their pivotal roles in the development and progression of cancer, making them promising targets for drug development. This article delves into multiple notable moonlighting enzymes, including GSK-3, GAPDH, and ENO1, and with a particular emphasis on an enigmatic phosphatase, PTP4A3. We scrutinize their distinct roles in cancer and the mechanisms that dictate their ability to switch roles. Lastly, we discuss the potential of an innovative approach to develop drugs targeting these moonlighting enzymes: target protein degradation. This strategy holds promise for effectively tackling moonlighting enzymes in the context of cancer therapy.
Collapse
Affiliation(s)
- Chunxu Lin
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Mingyang Yu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Ximei Wu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Hui Wang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Min Wei
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
| | - Luyong Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China; (C.L.); (M.Y.); (X.W.); (H.W.)
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
25
|
Mustafa M, Abbas K, Alam M, Ahmad W, Moinuddin, Usmani N, Siddiqui SA, Habib S. Molecular pathways and therapeutic targets linked to triple-negative breast cancer (TNBC). Mol Cell Biochem 2024; 479:895-913. [PMID: 37247161 DOI: 10.1007/s11010-023-04772-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/18/2023] [Indexed: 05/30/2023]
Abstract
Cancer is a group of diseases characterized by uncontrolled cellular growth, abnormal morphology, and altered proliferation. Cancerous cells lose their ability to act as anchors, allowing them to spread throughout the body and infiltrate nearby cells, tissues, and organs. If these cells are not identified and treated promptly, they will likely spread. Around 70% of female breast cancers are caused by a mutation in the BRCA gene, specifically BRCA1. The absence of progesterone, oestrogen and HER2 receptors (human epidermal growth factor) distinguishes the TNBC subtype of breast cancer. There were approximately 6,85,000 deaths worldwide and 2.3 million new breast cancer cases in women in 2020. Breast cancer is the most common cancer globally, affecting 7.8 million people at the end of 2020. Compared to other cancer types, breast cancer causes more women to lose disability-adjusted life years (DALYs). Worldwide, women can develop breast cancer at any age after puberty, but rates increase with age. The maintenance of mammary stem cell stemness is disrupted in TNBC, governed by signalling cascades controlling healthy mammary gland growth and development. Interpreting these essential cascades may facilitate an in-depth understanding of TNBC cancer and the search for an appropriate therapeutic target. Its treatment remains challenging because it lacks specific receptors, which renders hormone therapy and medications ineffective. In addition to radiotherapy, numerous recognized chemotherapeutic medicines are available as inhibitors of signalling pathways, while others are currently undergoing clinical trials. This article summarizes the vital druggable targets, therapeutic approaches, and strategies associated with TNBC.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Kashif Abbas
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Moinuddin
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India
| | - Nazura Usmani
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Shahid Ali Siddiqui
- Department of Radiotherapy, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
26
|
Schöckel L, Woischke C, Surendran SA, Michl M, Schiergens T, Hölscher A, Glass F, Kreissl P, Klauschen F, Günther M, Ormanns S, Neumann J. PPARG activation promotes the proliferation of colorectal cancer cell lines and enhances the antiproliferative effect of 5-fluorouracil. BMC Cancer 2024; 24:234. [PMID: 38378472 PMCID: PMC10877928 DOI: 10.1186/s12885-024-11985-5] [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: 10/09/2023] [Accepted: 02/08/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPARG) is a member of the nuclear receptor family. It is involved in the regulation of adipogenesis, lipid metabolism, insulin sensitivity, vascular homeostasis and inflammation. In addition, PPARG agonists, known as thiazolidinediones, are well established in the treatment of type 2 diabetes mellitus. PPARGs role in cancer is a matter of debate, as pro- and anti-tumour properties have been described in various tumour entities. Currently, the specific role of PPARG in patients with colorectal cancer (CRC) is not fully understood. MATERIAL AND METHODS The prognostic impact of PPARG expression was investigated by immunohistochemistry in a case-control study using a matched pair selection of CRC tumours (n = 246) with either distant metastases to the liver (n = 82), lung (n = 82) or without distant metastases (n = 82). Its effect on proliferation as well as the sensitivity to the chemotherapeutic drug 5-fluorouracil (5-FU) was examined after activation, inhibition, and transient gene knockdown of PPARG in the CRC cell lines SW403 and HT29. RESULTS High PPARG expression was significantly associated with pulmonary metastasis (p = 0.019). Patients without distant metastases had a significantly longer overall survival with low PPARG expression in their tumours compared to patients with high PPARG expression (p = 0.045). In the pulmonary metastasis cohort instead, a trend towards longer survival was observed for patients with high PPARG expression in their tumour (p = 0.059). Activation of PPARG by pioglitazone and rosiglitazone resulted in a significant dose-dependent increase in proliferation of CRC cell lines. Inhibition of PPARG by its specific inhibitor GW9662 and siRNA-mediated knockdown of PPARG significantly decreased proliferation. Activating PPARG significantly increased the CRC cell lines sensitivity to 5-FU while its inhibition decreased it. CONCLUSION The prognostic effect of PPARG expression depends on the metastasis localization in advanced CRC patients. Activation of PPARG increased malignancy associated traits such as proliferation in CRC cell lines but also increases sensitivity towards the chemotherapeutic agent 5-FU. Based on this finding, a combination therapy of PPARG agonists and 5-FU-based chemotherapy constitutes a promising strategy which should be further investigated.
Collapse
Affiliation(s)
- Leah Schöckel
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
| | - Christine Woischke
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
| | - Sai Agash Surendran
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
| | - Marlies Michl
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Tobias Schiergens
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | | | | | | | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU Munich Germany, Munich, Germany
| | - Michael Günther
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU Munich Germany, Munich, Germany
- Innpath Institute for Pathology GmbH, Tirol Kliniken, Innsbruck, Austria
| | - Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU Munich Germany, Munich, Germany
- Innpath Institute for Pathology GmbH, Tirol Kliniken, Innsbruck, Austria
| | - Jens Neumann
- Institute of Pathology, Ludwig-Maximilians-University (LMU) München, Munich, Germany.
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and LMU Munich Germany, Munich, Germany.
| |
Collapse
|
27
|
Schumann P, Rivetti C, Houghton J, Campos B, Hodges G, LaLone C. Combination of computational new approach methodologies for enhancing evidence of biological pathway conservation across species. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 912:168573. [PMID: 37981146 PMCID: PMC10926110 DOI: 10.1016/j.scitotenv.2023.168573] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
The ability to predict which chemicals are of concern for environmental safety is dependent, in part, on the ability to extrapolate chemical effects across many species. This work investigated the complementary use of two computational new approach methodologies to support cross-species predictions of chemical susceptibility: the US Environmental Protection Agency Sequence Alignment to Predict Across Species Susceptibility (SeqAPASS) tool and Unilever's recently developed Genes to Pathways - Species Conservation Analysis (G2P-SCAN) tool. These stand-alone tools rely on existing biological knowledge to help understand chemical susceptibility and biological pathway conservation across species. The utility and challenges of these combined computational approaches were demonstrated using case examples focused on chemical interactions with peroxisome proliferator activated receptor alpha (PPARα), estrogen receptor 1 (ESR1), and gamma-aminobutyric acid type A receptor subunit alpha (GABRA1). Overall, the biological pathway information enhanced the weight of evidence to support cross-species susceptibility predictions. Through comparisons of relevant molecular and functional data gleaned from adverse outcome pathways (AOPs) to mapped biological pathways, it was possible to gain a toxicological context for various chemical-protein interactions. The information gained through this computational approach could ultimately inform chemical safety assessments by enhancing cross-species predictions of chemical susceptibility. It could also help fulfill a core objective of the AOP framework by potentially expanding the biologically plausible taxonomic domain of applicability of relevant AOPs.
Collapse
Affiliation(s)
- Peter Schumann
- Oak Ridge Institute for Science and Education, Duluth, MN, USA
| | - Claudia Rivetti
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Jade Houghton
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Bruno Campos
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Geoff Hodges
- Safety and Environmental Assurance Centre, Unilever, Colworth Science Park, Sharnbrook, Bedfordshire, UK
| | - Carlie LaLone
- U.S. Environmental Protection Agency, Office of Research and Development, Center for Computational Toxicology and Exposure, Great Lakes Toxicology and Ecology Division, Duluth, MN, USA.
| |
Collapse
|
28
|
Peña-Oyarzún D, Flores T, Torres VA, Quest AFG, Lobos-González L, Kretschmar C, Contreras P, Maturana-Ramírez A, Criollo A, Reyes M. Inhibition of PORCN Blocks Wnt Signaling to Attenuate Progression of Oral Carcinogenesis. Clin Cancer Res 2024; 30:209-223. [PMID: 37812478 DOI: 10.1158/1078-0432.ccr-23-0318] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/12/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC) is commonly preceded by potentially malignant lesions, referred to as oral dysplasia. We recently reported that oral dysplasia is associated with aberrant activation of the Wnt/β-catenin pathway, due to overexpression of Wnt ligands in a Porcupine (PORCN)-dependent manner. Pharmacologic inhibition of PORCN precludes Wnt secretion and has been proposed as a potential therapeutic approach to treat established cancers. Nevertheless, there are no studies that explore the effects of PORCN inhibition at the different stages of oral carcinogenesis. EXPERIMENTAL DESIGN We performed a model of tobacco-induced oral cancer in vitro, where dysplastic oral keratinocytes (DOK) were transformed into oral carcinoma cells (DOK-TC), and assessed the effects of inhibiting PORCN with the C59 inhibitor. Similarly, an in vivo model of oral carcinogenesis and ex vivo samples derived from patients diagnosed with oral dysplasia and OSCC were treated with C59. RESULTS Both in vitro and ex vivo oral carcinogenesis approaches revealed decreased levels of nuclear β-catenin and Wnt3a, as observed by immunofluorescence and IHC analyses. Consistently, reduced protein and mRNA levels of survivin were observed after treatment with C59. Functionally, treatment with C59 in vitro resulted in diminished cell migration, viability, and invasion. Finally, by using an in vivo model of oral carcinogenesis, we found that treatment with C59 prevented the development of OSCC by reducing the size and number of oral tumor lesions. CONCLUSIONS The inhibition of Wnt ligand secretion with C59 represents a feasible treatment to prevent the progression of early oral lesions toward OSCC.
Collapse
Affiliation(s)
- Daniel Peña-Oyarzún
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Physiology Department, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Interdisciplinary Center for Research in Territorial Health of the Aconcagua Valley (CIISTe Aconcagua), School of Medicine, Faculty of Medicine, San Felipe Campus, Universidad de Valparaiso, Chile
| | - Tania Flores
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Research Centre in Dental Science (CICO), Faculty of Dentistry, Universidad de La Frontera, Temuco, Chile
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Vicente A Torres
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Center for Regenerative Medicine, Faculty of Medicine, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Catalina Kretschmar
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Pamela Contreras
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Laboratory of Cellular Communication, Center for studies on Exercise, Metabolism and Cancer (CEMC), Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Andrea Maturana-Ramírez
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alfredo Criollo
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Montserrat Reyes
- Department of Pathology and Oral Medicine, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
29
|
Zheng C, Huang J, Xu G, Li W, Weng X, Zhang S. The Notch signaling pathway in desmoid tumor: Recent advances and the therapeutic prospects. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166907. [PMID: 37793461 DOI: 10.1016/j.bbadis.2023.166907] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023]
Abstract
Desmoid tumor (DT) is a rare fibroblastic soft-tissue neoplasm that is characterized by local aggressiveness but no metastatic potential. Although the prognosis is relatively favorable, the unpredictable disease course and infiltrative growth lead to significant impairments and morbidity. Aberrant activation of Wnt/β-catenin signaling has been well-established in the pathogenesis of sporadic DT and familial adenomatous polyposis (FAP) or Gardners syndrome-associated DT, suggesting therapy targeting this pathway is an appealing treatment strategy. However, agents against this pathway are currently in their preliminary stages and have not yet been implemented in clinical practice. Increasing studies demonstrate activation of the Notch pathway is closely associated with the development and progression of DT, which provides a potential alternative therapeutic target against DT. Early-stage clinical trials and preclinical models have indicated that inhibition of Notch pathway might be a promising treatment approach for DT. The Notch signaling activation is mainly dependent on the activity of the γ-secretase enzyme, which is responsible for cleaving the Notch intracellular domain and facilitating its nuclear translocation to promote gene transcription. Two γ-secretase inhibitors called nirogacestat and AL102 are currently under extensive investigation in the advanced stage of clinical development. The updated findings from the phase III randomized controlled trial (DeFi trial) demonstrated that nirogacestat exerts significant benefits in terms of disease control and symptom resolution in patients with progressive DT. Therefore, this review provides a comprehensive overview of the present understanding of Notch signaling in the pathogenesis of DT, with a particular emphasis on the prospective therapeutic application of γ-secretase inhibitors in the management of DT.
Collapse
Affiliation(s)
- Chuanxi Zheng
- Department of Musculoskeletal Tumor Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Jianghong Huang
- Department of Spine Surgery and Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen 518035, China
| | - Gang Xu
- Department of Musculoskeletal Tumor Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Wei Li
- Department of Musculoskeletal Tumor Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Xin Weng
- Department of Pathology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Shiquan Zhang
- Department of Musculoskeletal Tumor Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| |
Collapse
|
30
|
Shi K, Shan Y, Sun X, Chen K, Luo Q, Xu Q. TP53INP2 modulates the malignant progression of colorectal cancer by reducing the inactive form of β-catenin. Biochem Biophys Res Commun 2024; 690:149275. [PMID: 37995453 DOI: 10.1016/j.bbrc.2023.149275] [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: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
TP53INP2 (tumor protein p53-inducible nuclear protein 2), known as an autophagy protein, is essential for regulating transcription and starvation-induced autophagy, which plays a crucial role in the oncogenesis and progression of various cancers. The present study aims to investigate the expression pattern, function and prognostic value of TP53INP2 in colorectal cancer (CRC). Here, we report that low expression of TP53INP2 correlates with poor survival in CRC patients. TP53INP2 was significantly downregulated in CRC tissues compared with adjacent tissues. As the malignancy of CRC progresses, the expression level of TP53INP2 gradually decreased. Knockdown of TP53INP2 promoted CRC cell proliferation and tumor growth in mice. Mechanistically, TP53INP2 deficiency decreased phosphorylation of β-catenin on S33, S37, and T41, resulting in increased accumulation of β-catenin and enhanced nuclear translocation and transcriptional activity. Moreover, we further demonstrated that TP53INP2 sequestered TIM50, thereby inhibiting its activation of β-catenin. Taken together, our findings indicate that the downregulation of TP53INP2 promotes CRC progression by activating β-catenin and suggest that TP53INP2 may be a candidate therapeutic target for CRC.
Collapse
Affiliation(s)
- Ke Shi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiao Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Kuida Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China
| | - Qiong Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
31
|
Dev A, Vachher M, Prasad CP. β-catenin inhibitors in cancer therapeutics: intricacies and way forward. Bioengineered 2023; 14:2251696. [PMID: 37655825 PMCID: PMC10478749 DOI: 10.1080/21655979.2023.2251696] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023] Open
Abstract
β-catenin is an evolutionary conserved, quintessential, multifaceted protein that plays vital roles in cellular homeostasis, embryonic development, organogenesis, stem cell maintenance, cell proliferation, migration, differentiation, apoptosis, and pathogenesis of various human diseases including cancer. β-catenin manifests both signaling and adhesive features. It acts as a pivotal player in intracellular signaling as a component of versatile WNT signaling cascade involved in embryonic development, homeostasis as well as in carcinogenesis. It is also involved in Ca2+ dependent cell adhesion via interaction with E-cadherin at the adherens junctions. Aberrant β-catenin expression and its nuclear accumulation promote the transcription of various oncogenes including c-Myc and cyclinD1, thereby contributing to tumor initiation, development, and progression. β-catenin's expression is closely regulated at various levels including its stability, sub-cellular localization, as well as transcriptional activity. Understanding the molecular mechanisms of regulation of β-catenin and its atypical expression will provide researchers not only the novel insights into the pathogenesis and progression of cancer but also will help in deciphering new therapeutic avenues. In the present review, we have summarized the dual functions of β-catenin, its role in signaling, associated mutations as well as its role in carcinogenesis and tumor progression of various cancers. Additionally, we have discussed the challenges associated with targeting β-catenin molecule with the presently available drugs and suggested the possible way forward in designing new therapeutic alternatives against this oncogene.
Collapse
Affiliation(s)
- Arundhathi Dev
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Meenakshi Vachher
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
32
|
Iyer D, Mastrogiacomo DM, Li K, Banerjee R, Yang Y, Scallan JP. eNOS Regulates Lymphatic Valve Specification by Controlling β-Catenin Signaling During Embryogenesis in Mice. Arterioscler Thromb Vasc Biol 2023; 43:2197-2212. [PMID: 37767708 PMCID: PMC10655861 DOI: 10.1161/atvbaha.123.319405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Lymphatic valves play a critical role in ensuring unidirectional lymph transport. Loss of lymphatic valves or dysfunctional valves are associated with several diseases including lymphedema, lymphatic malformations, obesity, and ileitis. Lymphatic valves first develop during embryogenesis in response to mechanotransduction signaling pathways triggered by oscillatory lymph flow. In blood vessels, eNOS (endothelial NO synthase; gene name: Nos3) is a well-characterized shear stress signaling effector, but its role in lymphatic valve development remains unexplored. METHODS We used global Nos3-/- mice and cultured human dermal lymphatic endothelial cells to investigate the role of eNOS in lymphatic valve development, which requires oscillatory shear stress signaling. RESULTS Our data reveal a 45% reduction in lymphatic valve specification cell clusters and that loss of eNOS protein inhibited activation of β-catenin and its nuclear translocation. Genetic knockout or knockdown of eNOS led to downregulation of β-catenin target proteins in vivo and in vitro. However, pharmacological inhibition of NO production did not reproduce these effects. Co-immunoprecipitation and proximity ligation assays reveal that eNOS directly binds to β-catenin and their binding is enhanced by oscillatory shear stress. Finally, genetic ablation of the Foxo1 gene enhanced FOXC2 expression and partially rescued the loss of valve specification in the eNOS knockouts. CONCLUSIONS In conclusion, we demonstrate a novel, NO-independent role for eNOS in regulating lymphatic valve specification and propose a mechanism by which eNOS directly binds β-catenin to regulate its nuclear translocation and thereby transcriptional activity.
Collapse
Affiliation(s)
- Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Diandra M Mastrogiacomo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Kunyu Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
33
|
Zhou RW, Harpaz N, Itzkowitz SH, Parsons RE. Molecular mechanisms in colitis-associated colorectal cancer. Oncogenesis 2023; 12:48. [PMID: 37884500 PMCID: PMC10603140 DOI: 10.1038/s41389-023-00492-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 10/28/2023] Open
Abstract
Sustained chronic inflammation of the large intestine leads to tissue damage and repair, which is associated with an increased incidence of colitis-associated colorectal cancer (CAC). The genetic makeup of CAC is somewhat similar to sporadic colorectal carcinoma (sCRC), but there are differences in the sequence and timing of alterations in the carcinogenesis process. Several models have been developed to explain the development of CAC, particularly the "field cancerization" model, which proposes that chronic inflammation accelerates mutagenesis and selects for the clonal expansion of phenotypically normal, pro-tumorigenic cells. In contrast, the "Big Bang" model posits that tumorigenic clones with multiple driver gene mutations emerge spontaneously. The details of CAC tumorigenesis-and how they differ from sCRC-are not yet fully understood. In this Review, we discuss recent genetic, epigenetic, and environmental findings related to CAC pathogenesis in the past five years, with a focus on unbiased, high-resolution genetic profiling of non-dysplastic field cancerization in the context of inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Royce W Zhou
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Molecular Medicine Program, Internal Medicine Residency Program, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Noam Harpaz
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven H Itzkowitz
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ramon E Parsons
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Hassan SA, Shabaan AAA, Ahmed AR, Issa YA, Fadel SH, El-Sabaa BM. Clinicopathological significance of SOX9 and β-catenin expression in pre-neoadjuvant chemotherapy cases of osteosarcoma: molecular and immunohistochemical study. J Histotechnol 2023; 46:127-138. [PMID: 37013797 DOI: 10.1080/01478885.2023.2193526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
The molecular pathogenesis of osteosarcoma (OS), the most frequent primary malignant bone tumor of all age groups, is still obscure. Since multidrug chemotherapeutic regimens were introduced in the 1970s, survival rates have been stationary. The Wnt-β-catenin signaling cascade and SOX9 have a significant contribution to skeletal growth, development, and tumorigenesis. In the present work, an attempt was made to examine the role and clinicopathological significance of β-catenin and SOX9 in 46 cases of pre-neoadjuvant chemotherapy OS tissues compared to 10 cases of non-neoplastic bone. The mRNA levels of both markers were assessed by qRT-PCR, and protein levels of β-catenin were analyzed by immunohistochemistry. The results were correlated with different clinicopathological parameters. SOX9 mRNA levels were significantly elevated in OS compared to non-neoplastic bone, and higher levels were significantly associated with the occurrence of fluid-fluid levels (indicating blood-containing cystic spaces) and osteolytic radiological pattern. Although β-catenin mRNA and protein levels were higher in OS compared to non-neoplastic bone, only the protein levels reached statistical significance. Higher β-catenin mRNA levels were significantly associated with tumor size, while higher protein levels were significantly associated with the histologic subtype, mitotic count, and radiological pattern. No significant association was noted with any of the other evaluated parameters. OS showing higher SOX9 mRNA expression and lower β-catenin mRNA and protein expression exhibited longer estimated overall survival times approaching statistical significance. To conclude, while high expression of β-catenin and SOX9 suggests their possible involvement in OS development, their prognostic role may need further research.
Collapse
Affiliation(s)
- Sarah Ahmed Hassan
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Adel Refaat Ahmed
- Department of Orthopedic Surgery and Traumatology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Yasmine Amr Issa
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Shady Hassan Fadel
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Faculty of Medicine, Alexandria, Egypt
| | | |
Collapse
|
35
|
Yu Y, Papukashvili D, Ren R, Rcheulishvili N, Feng S, Bai W, Zhang H, Xi Y, Lu X, Xing N. siRNA-based approaches for castration-resistant prostate cancer therapy targeting the androgen receptor signaling pathway. Future Oncol 2023; 19:2055-2073. [PMID: 37823367 DOI: 10.2217/fon-2023-0227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
Androgen deprivation therapy is a common treatment method for metastatic prostate cancer through lowering androgen levels; however, this therapy frequently leads to the development of castration-resistant prostate cancer (CRPC). This is attributed to the activation of the androgen receptor (AR) signaling pathway. Current treatments targeting AR are often ineffective mostly due to AR gene overexpression and mutations, as well as the presence of splice variants that accelerate CRPC progression. Thus there is a critical need for more specific medication to treat CRPC. Small interfering RNAs have shown great potential as a targeted therapy. This review discusses prostate cancer progression and the role of AR signaling in CRPC, and proposes siRNA-based targeted therapy as a promising strategy for CRPC.
Collapse
Affiliation(s)
- Yanling Yu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | | | - Ruimin Ren
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Urology, Taiyuan, 030032, China
| | | | - Shunping Feng
- Southern University of Science & Technology, Shenzhen, 518000, China
| | - Wenqi Bai
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Huanhu Zhang
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Yanfeng Xi
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaoqing Lu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Nianzeng Xing
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
36
|
Jiang F, Du L, Chen ZJ, Wang X, Ge D, Liu N. LNP-miR-155 cy5 Inhibitor Regulates the Copper Transporter via the β-Catenin/TCF4/SLC31A1 Signal for Colorectal Cancer Therapy. Mol Pharm 2023; 20:4138-4152. [PMID: 37358225 DOI: 10.1021/acs.molpharmaceut.3c00276] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Lipid nanoparticle (LNP) delivery systems are widely used in the delivery of small-molecule drugs and nucleic acids. In this study, we prepared LNP-miR-155 by lipid nanomaterial technology and investigated the effects of LNP-miR-155 on β-catenin/transcription factor 4 (TCF4)/solute carrier family 31 member 1/copper transporter 1 (SLC31A1/CTR1) signaling and copper transport in colorectal cancer. For this, we used an LNP-miR-155 cy5 inhibitor and LNP-miR-155 cy5 mimics for the transfection of HT-29/SW480 cells. The transfection efficiency and uptake efficiency were detected by immunofluorescence. Relevant cell assays confirmed that the LNP-miR-155 cy5 inhibitor mediates the regulation of copper transport through the β-catenin/TCF4/SLC31A1 axis. The LNP-miR-155 cy5 inhibitor reduced cell proliferation, migration, and colony formation and promoted cell apoptosis. We also confirmed that miR-155 downregulates HMG box-containing protein 1 (HBP1) and adenomatous polyposis coli (APC) in cells and activates the function of β-catenin/TCF4 signaling. In addition, we found that the copper transporter, SLC31A1, is highly expressed in colorectal cancer cells. Furthermore, we also found that the complex β-catenin/TCF4 promotes the transcription of SLC31A1 by binding to its promoter region, which sustains the transport of copper from the extracellular region to the intracellular region and increases the activities of Cu2+-ATPase and superoxide dismutase (SOD). In summary, the LNP-miR-155 cy5 inhibitor regulates β-catenin/TCF4 by downregulating SLC31A1-mediated copper transport and intracellular copper homeostasis.
Collapse
Affiliation(s)
- Fan Jiang
- Department of the Center of Gerontology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P. R. China
| | - Le Du
- Department of Biology, Hainan Medical University, Haikou, Hainan 570100, P. R. China
| | - Zhi-Ju Chen
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P. R. China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P. R. China
| | - Dongsheng Ge
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P. R. China
| | - Ning Liu
- Department of Gastrointestinal Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P. R. China
| |
Collapse
|
37
|
Papukashvili D, Liu C, Rcheulishvili N, Xie F, Wang X, Feng S, Sun X, Zhang C, Li Y, He Y, Wang PG. DKK1-targeting cholesterol-modified siRNA implication in hair growth regulation. Biochem Biophys Res Commun 2023; 668:55-61. [PMID: 37244035 DOI: 10.1016/j.bbrc.2023.05.047] [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: 04/16/2023] [Revised: 05/03/2023] [Accepted: 05/14/2023] [Indexed: 05/29/2023]
Abstract
Despite advancements in medical research, androgenetic alopecia (AGA) remains a humankind problem that still needs to be overcome. To date, clinical practice lacks an ideal treatment for AGA. The Wnt/β-catenin signaling pathway is evidenced to play a key role in hair regrowth, hence, modulating this signaling pathway for AGA therapy appears to be rational. One of the major inhibitors of the canonical Wnt/β-catenin signaling pathway is dickkopf-related protein 1 (DKK1). In this report, we have selected a small interfering RNA (siRNA) targeting DKK1 in vitro via qPCR and then tested its efficacy in vivo on the depilated dorsal skin of the mice. The changes in hair growth in different groups were observed over time. Moreover, the visual observation of the hair growth and hematoxylin and eosin (HE) staining showed that DKK1-targeting siRNA reveals non-inferior results compared with the mice treated with the Food and Drug Administration (FDA)-approved, commercially available minoxidil (5%) topical solution that was used as a positive control. Both- positive control and DKK1-targeting siRNA groups demonstrated significantly superior results compared with the control group that received negative control siRNA. Consequently, siRNAs targeting DKK1 may promote hair growth regulation in the AGA population via potentially activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Dimitri Papukashvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Cong Liu
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Nino Rcheulishvili
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Fengfei Xie
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Xingyun Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Shunping Feng
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Xiu Sun
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Chi Zhang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Yingyu Li
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Yunjiao He
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China.
| | - Peng George Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
38
|
Sharma T, Olea-Flores M, Imbalzano AN. Regulation of the Wnt signaling pathway during myogenesis by the mammalian SWI/SNF ATPase BRG1. Front Cell Dev Biol 2023; 11:1160227. [PMID: 37484913 PMCID: PMC10360407 DOI: 10.3389/fcell.2023.1160227] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Skeletal muscle differentiation is a tightly regulated process, and the importance of the mammalian SWI/SNF (mSWI/SNF) chromatin remodeling family for regulation of genes involved in skeletal myogenesis is well-established. Our prior work showed that bromodomains of mSWI/SNF ATPases BRG1 and BRM contribute to myogenesis by facilitating the binding of mSWI/SNF enzymes to regulatory regions of myogenic and other target genes. Here, we report that pathway analyses of differentially expressed genes from that study identified an additional role for mSWI/SNF enzymes via the regulation of the Wnt signaling pathway. The Wnt pathway has been previously shown to be important for skeletal muscle development. To investigate the importance of mSWI/SNF enzymes for the regulation of the Wnt pathway, individual and dual knockdowns were performed for BRG1 and BRM followed by RNA-sequencing. The results show that BRG1, but not BRM, is a regulator of Wnt pathway components and downstream genes. Reactivation of Wnt pathway by stabilization of β-catenin could rescue the defect in myogenic gene expression and differentiation due to BRG1 knockdown or bromodomain inhibition using a specific small molecule inhibitor, PFI-3. These results demonstrate that BRG1 is required upstream of β-catenin function. Chromatin immunoprecipitation of BRG1, BRM and β-catenin at promoters of Wnt pathway component genes showed binding of BRG1 and β-catenin, which provides further mechanistic insight to the transcriptional regulation of these genes.
Collapse
Affiliation(s)
| | | | - Anthony N. Imbalzano
- Department of Biochemistry and Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, United States
| |
Collapse
|
39
|
Iyer D, Mastrogiacomo D, Li K, Banerjee R, Yang Y, Scallan JP. Endothelial Nitric Oxide Synthase Regulates Lymphatic Valve Specification By Controlling β - catenin Signaling During Embryogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536303. [PMID: 37090551 PMCID: PMC10120724 DOI: 10.1101/2023.04.10.536303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Objective Lymphatic valves play a critical role in ensuring unidirectional lymph transport. Loss of lymphatic valves or dysfunctional valves are associated with several diseases including lymphedema, lymphatic malformations, obesity, and ileitis. Lymphatic valves first develop during embryogenesis in response to mechanotransduction signaling pathways triggered by oscillatory lymph flow. In blood vessels, eNOS (gene name: Nos3 ) is a well characterized shear stress signaling effector, but its role in lymphatic valve development remains unexplored. Approach and Results We used global Nos3 -/- mice and cultured hdLECs to investigate the role of eNOS in lymphatic valve development, which requires oscillatory shear stress signaling. Our data reveal a 45% reduction in lymphatic valve specification cell clusters and that loss of eNOS protein inhibited activation of β-catenin and its nuclear translocation. Genetic knockout or knockdown of eNOS led to downregulation of β-catenin target proteins in vivo and in vitro . However, pharmacological inhibition of NO production did not reproduce these effects. Coimmunoprecipitation experiments reveal that eNOS forms a complex with β-catenin and their association is enhanced by oscillatory shear stress. Finally, genetic ablation of the Foxo1 gene enhanced FOXC2 expression and rescued the loss of valve specification in the eNOS knockouts. Conclusion In conclusion, we demonstrate a novel, nitric oxide-independent role for eNOS in regulating lymphatic valve specification and propose a mechanism by which eNOS forms a complex with β-catenin to regulate its nuclear translocation and thereby transcriptional activity.
Collapse
Affiliation(s)
- Drishya Iyer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Diandra Mastrogiacomo
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Kunyu Li
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Richa Banerjee
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL USA 33612
| |
Collapse
|
40
|
Park WJ, Kim MJ. A New Wave of Targeting 'Undruggable' Wnt Signaling for Cancer Therapy: Challenges and Opportunities. Cells 2023; 12:cells12081110. [PMID: 37190019 DOI: 10.3390/cells12081110] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Aberrant Wnt signaling activation is frequently observed in many cancers. The mutation acquisition of Wnt signaling leads to tumorigenesis, whereas the inhibition of Wnt signaling robustly suppresses tumor development in various in vivo models. Based on the excellent preclinical effect of targeting Wnt signaling, over the past 40 years, numerous Wnt-targeted therapies have been investigated for cancer treatment. However, Wnt signaling-targeting drugs are still not clinically available. A major obstacle to Wnt targeting is the concomitant side effects during treatment due to the pleiotropic role of Wnt signaling in development, tissue homeostasis, and stem cells. Additionally, the complexity of the Wnt signaling cascades across different cancer contexts hinders the development of optimized targeted therapies. Although the therapeutic targeting of Wnt signaling remains challenging, alternative strategies have been continuously developed alongside technological advances. In this review, we give an overview of current Wnt targeting strategies and discuss recent promising trials that have the potential to be clinically realized based on their mechanism of action. Furthermore, we highlight new waves of Wnt targeting that combine recently developed technologies such as PROTAC/molecular glue, antibody-drug conjugates (ADC), and anti-sense oligonucleotides (ASO), which may provide us with new opportunities to target 'undruggable' Wnt signaling.
Collapse
Affiliation(s)
- Woo-Jung Park
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Moon Jong Kim
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Lee Gil Ya Cancer and Diabetes Institute, Incheon 21999, Republic of Korea
| |
Collapse
|
41
|
Palamarchuk AI, Kovalenko EI, Streltsova MA. Multiple Actions of Telomerase Reverse Transcriptase in Cell Death Regulation. Biomedicines 2023; 11:biomedicines11041091. [PMID: 37189709 DOI: 10.3390/biomedicines11041091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/25/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Telomerase reverse transcriptase (TERT), a core part of telomerase, has been known for a long time only for its telomere lengthening function by reverse transcription of RNA template. Currently, TERT is considered as an intriguing link between multiple signaling pathways. The diverse intracellular localization of TERT corresponds to a wide range of functional activities. In addition to the canonical function of protecting chromosome ends, TERT by itself or as a part of the telomerase complex participates in cell stress responses, gene regulation and mitochondria functioning. Upregulation of TERT expression and increased telomerase activity in cancer and somatic cells relate to improved survival and persistence of such cells. In this review, we summarize the data for a comprehensive understanding of the role of TERT in cell death regulation, with a focus on the interaction of TERT with signaling pathways involved in cell survival and stress response.
Collapse
Affiliation(s)
- Anastasia I. Palamarchuk
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Elena I. Kovalenko
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Maria A. Streltsova
- Shemyakin & Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
42
|
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
|
43
|
Kaiser A, Eiselt G, Bechler J, Huber O, Schmidt M. WNT3a Signaling Inhibits Aromatase Expression in Breast Adipose Fibroblasts-A Possible Mechanism Supporting the Loss of Estrogen Responsiveness of Triple-Negative Breast Cancers. Int J Mol Sci 2023; 24:ijms24054654. [PMID: 36902090 PMCID: PMC10003471 DOI: 10.3390/ijms24054654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Estrogen-dependent breast cancers rely on a constant supply of estrogens and expression of estrogen receptors. Local biosynthesis, by aromatase in breast adipose fibroblasts (BAFs), is their most important source for estrogens. Triple-negative breast cancers (TNBC) rely on other growth-promoting signals, including those from the Wnt pathway. In this study, we explored the hypothesis that Wnt signaling alters the proliferation of BAFs, and is involved in regulation of aromatase expression in BAFs. Conditioned medium (CM) from TNBC cells and WNT3a consistently increased BAF growth, and reduced aromatase activity up to 90%, by suppression of the aromatase promoter I.3/II region. Database searches identified three putative Wnt-responsive elements (WREs) in the aromatase promoter I.3/II. In luciferase reporter gene assays, promoter I.3/II activity was inhibited by overexpression of full-length T-cell factor (TCF)-4 in 3T3-L1 preadipocytes, which served as a model for BAFs. Full-length lymphoid enhancer-binding factor (LEF)-1 increased the transcriptional activity. However, TCF-4 binding to WRE1 in the aromatase promoter, was lost after WNT3a stimulation in immunoprecipitation-based in vitro DNA-binding assays, and in chromatin immunoprecipitation (ChIP). In vitro DNA-binding assays, ChIP, and Western blotting revealed a WNT3a-dependent switch of nuclear LEF-1 isoforms towards a truncated variant, whereas β-catenin levels remained unchanged. This LEF-1 variant revealed dominant negative properties, and most likely recruited enzymes involved in heterochromatin formation. In addition, WNT3a induced the replacement of TCF-4 by the truncated LEF-1 variant, on WRE1 of the aromatase promoter I.3/II. The mechanism described here may be responsible for the loss of aromatase expression predominantly associated with TNBC. Tumors with (strong) expression of Wnt ligands actively suppress aromatase expression in BAFs. Consequently a reduced estrogen supply could favor the growth of estrogen-independent tumor cells, which consequently would make estrogen receptors dispensable. In summary, canonical Wnt signaling within (cancerous) breast tissue may be a major factor controlling local estrogen synthesis and action.
Collapse
Affiliation(s)
- Alexander Kaiser
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Gabriele Eiselt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Joachim Bechler
- Department of Gynecology and Obstetrics, Robert-Koch-Hospital, 99510 Apolda, Germany
| | - Otmar Huber
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-939-6420
| |
Collapse
|
44
|
Zhao X, Richardson DR. The role of the NDRG1 in the pathogenesis and treatment of breast cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188871. [PMID: 36841367 DOI: 10.1016/j.bbcan.2023.188871] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women. This disease is heterogeneous, with clinical subtypes being estrogen receptor-α (ER-α) positive, having human epidermal growth factor receptor 2 (HER2) overexpression, or being triple-negative for ER-α, progesterone receptor, and HER2 (TNBC). The ER-α positive and HER2 overexpressing tumors can be treated with agents targeting these proteins, including tamoxifen and pertuzumab, respectively. Despite these treatments, resistance and metastasis are problematic, while TNBC is challenging to treat due to the lack of suitable targets. Many studies examining BC and other tumors indicate a role for N-myc downstream-regulated gene-1 (NDRG1) as a metastasis suppressor. The ability of NDRG1 to inhibit metastasis is due, in part, to the inhibition of the initial step in metastasis, namely the epithelial-to-mesenchymal transition. Paradoxically, there are also reports of NDRG1 playing a pro-oncogenic role in BC pathogenesis. The oncogenic effects of NDRG1 in BC have been reported to relate to lipid metabolism or the mTOR signaling pathway. The molecular mechanism(s) of how NDRG1 regulates the activity of multiple signaling pathways remains unclear. Therapeutic strategies that up-regulate NDRG1 have been developed and include agents of the di-2-pyridylketone thiosemicarbazone class. These compounds target oncogenic drivers in BC cells, suppressing the expression of multiple key hormone receptors including ER-α, progesterone receptor, androgen receptor, and prolactin receptor, and can also overcome tamoxifen resistance. Considering the varying role of NDRG1 in BC pathogenesis, further studies are required to examine what subset of BC patients would benefit from pharmacopeia that up-regulate NDRG1.
Collapse
Affiliation(s)
- Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
45
|
Schäfer J, Wenck N, Janik K, Linnert J, Stingl K, Kohl S, Nagel-Wolfrum K, Wolfrum U. The Usher syndrome 1C protein harmonin regulates canonical Wnt signaling. Front Cell Dev Biol 2023; 11:1130058. [PMID: 36846582 PMCID: PMC9944737 DOI: 10.3389/fcell.2023.1130058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Human Usher syndrome (USH) is the most common form of hereditary combined deaf-blindness. USH is a complex genetic disorder, and the pathomechanisms underlying the disease are far from being understood, especially in the eye and retina. The USH1C gene encodes the scaffold protein harmonin which organizes protein networks due to binary interactions with other proteins, such as all USH proteins. Interestingly, only the retina and inner ear show a disease-related phenotype, although USH1C/harmonin is almost ubiquitously expressed in the human body and upregulated in colorectal cancer. We show that harmonin binds to β-catenin, the key effector of the canonical Wnt (cWnt) signaling pathway. We also demonstrate the interaction of the scaffold protein USH1C/harmonin with the stabilized acetylated β-catenin, especially in nuclei. In HEK293T cells, overexpression of USH1C/harmonin significantly reduced cWnt signaling, but a USH1C-R31* mutated form did not. Concordantly, we observed an increase in cWnt signaling in dermal fibroblasts derived from an USH1C R31*/R80Pfs*69 patient compared with healthy donor cells. RNAseq analysis reveals that both the expression of genes related to the cWnt signaling pathway and cWnt target genes were significantly altered in USH1C patient-derived fibroblasts compared to healthy donor cells. Finally, we show that the altered cWnt signaling was reverted in USH1C patient fibroblast cells by the application of Ataluren, a small molecule suitable to induce translational read-through of nonsense mutations, hereby restoring some USH1C expression. Our results demonstrate a cWnt signaling phenotype in USH establishing USH1C/harmonin as a suppressor of the cWnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jessica Schäfer
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nicole Wenck
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katharina Janik
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Joshua Linnert
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katarina Stingl
- Centre for Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Susanne Kohl
- Centre for Ophthalmology, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany,Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany,*Correspondence: Uwe Wolfrum,
| |
Collapse
|
46
|
Selvarathinam K, Subramani P, Thekkumalai M, Vilwanathan R, Selvarajan R, Abia ALK. Wnt Signaling Pathway Collapse upon β-Catenin Destruction by a Novel Antimicrobial Peptide SKACP003: Unveiling the Molecular Mechanism and Genetic Activities Using Breast Cancer Cell Lines. Molecules 2023; 28:molecules28030930. [PMID: 36770598 PMCID: PMC9920962 DOI: 10.3390/molecules28030930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Despite progress in breast cancer treatment, the survival rate for patients with metastatic breast cancer remains low due to chemotherapeutic agent resistance and the lack of specificity of the current generation of cancer drugs. Our previous findings indicated that the antimicrobial peptide SKACP003 exhibited anticancer properties, particularly against the MCF-7, MDA-MB-231, and MDA-MB-453 breast cancer cell lines. However, the mechanism of SKACP003-induced cancer cell death is unknown. Here, we investigated the molecular mechanism by which SKACP003 inhibits the cell cycle, cell proliferation, and angiogenesis in breast cancer cell lines. The results revealed that all the breast cancer cell lines treated at their IC50 values significantly inhibited the replicative phase of the cell cycle. The SKACP003-induced growth inhibition induced apoptosis, as evidenced by a decrease in BCL-2 and an increase in BAX and caspase gene (Cas-3, Cas-8, and Cas-9) expression. Reduced expression of the β-Catenin signaling pathway was associated with the SKACP003-induced apoptosis. SKACP003-treated breast cancer cells showed decreased expression of Wnt/β-Catenin targeting genes such as C-Myc, P68, and COX-2 and significant downregulation of CDK-4 and CDK-6 genes. Furthermore, cytoplasmic β-catenin protein levels in SKACP003-treated cell lines were significantly lower than in control cell lines. The results of the current study suggest that the newly identified antimicrobial peptide SKACP003 has great potential as a candidate for specifically targeting the β-catenin and thus significantly reducing the progression and prognosis of breast cancer cell lines.
Collapse
Affiliation(s)
- Kanitha Selvarathinam
- Department of Biochemistry, J.J. College of Arts and Science (Autonomous), Pudukkottai 622422, Tamilnadu, India
- Correspondence: (K.S.); (A.L.K.A.)
| | - Prabhu Subramani
- Department of Biochemistry, School of Life Science, Bharathidasan University, Tiruchirappalli 622422, Tamilnadu, India
| | | | - Ravikumar Vilwanathan
- Department of Biochemistry, School of Life Science, Bharathidasan University, Tiruchirappalli 622422, Tamilnadu, India
| | - Ramganesh Selvarajan
- Department of Environmental Sciences, College of Agricultural and Environmental Sciences (CAES), University of South Africa (UNISA), Florida—Campus, Florida Park, Roodepoort 1709, South Africa
- Laboratory of Extraterrestrial Ocean Systems (LEOS), Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences (CAS), Sanya 572000, China
| | - Akebe Luther King Abia
- Department of Environmental Sciences, College of Agricultural and Environmental Sciences (CAES), University of South Africa (UNISA), Florida—Campus, Florida Park, Roodepoort 1709, South Africa
- Environmental Research Foundation, Westville 3630, South Africa
- Correspondence: (K.S.); (A.L.K.A.)
| |
Collapse
|
47
|
Wang H, Zhao M, Shi F, Zheng S, Xiong L, Zheng L. A review of signal pathway induced by virulent protein CagA of Helicobacter pylori. Front Cell Infect Microbiol 2023; 13:1062803. [PMID: 37124036 PMCID: PMC10140366 DOI: 10.3389/fcimb.2023.1062803] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Gastric cancer (GC), a common and high-mortality disease, still occupies an important position in current cancer research, and Helicobacter pylori (H. pylori) infection as its important risk factor has been a hot and challenging research area. Among the numerous pathogenic factors of H. pylori, the virulence protein CagA has been widely studied as the only bacterial-derived oncoprotein. It was found that CagA entering into gastric epithelial cells (GECs) can induce the dysregulation of multiple cellular pathways such as MAPK signaling pathway, PI3K/Akt signaling pathway, NF-κB signaling pathway, Wnt/β-catenin signaling pathway, JAK-STAT signaling pathway, Hippo signaling pathway through phosphorylation and non-phosphorylation. These disordered pathways cause pathological changes in morphology, adhesion, polarity, proliferation, movement, and other processes of GECs, which eventually promotes the occurrence of GC. With the deepening of H. pylori-related research, the research on CagA-induced abnormal signaling pathway has been updated and deepened to some extent, so the key signaling pathways activated by CagA are used as the main stem to sort out the pathogenesis of CagA in this paper, aiming to provide new strategies for the H. pylori infection and treatment of GC in the future.
Collapse
Affiliation(s)
- Haiqiang Wang
- Department of Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mei Zhao
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fan Shi
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shudan Zheng
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Li Xiong
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lihong Zheng
- Department of Internal Medicine, Fourth Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Lihong Zheng,
| |
Collapse
|
48
|
Zhu X, Gao H, Qin S, Liu D, Cairns J, Gu Y, Yu J, Weinshilboum RM, Wang L. Testis- specific Y-encoded- like protein 1 and cholesterol metabolism: Regulation of CYP1B1 expression through Wnt signaling. Front Pharmacol 2022; 13:1047318. [PMID: 36518674 PMCID: PMC9742362 DOI: 10.3389/fphar.2022.1047318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/07/2022] [Indexed: 08/30/2023] Open
Abstract
The cytochromes P450 (CYPs) represent a large gene superfamily that plays an important role in the metabolism of both exogenous and endogenous compounds. We have reported that the testis-specific Y-encoded-like proteins (TSPYLs) are novel CYP gene transcriptional regulators. However, little is known of mechanism(s) by which TSPYLs regulate CYP expression or the functional consequences of that regulation. The TSPYL gene family includes six members, TSPYL1 to TSPYL6. However, TSPYL3 is a pseudogene, TSPYL5 is only known to regulates the expression of CYP19A1, and TSPYL6 is expressed exclusively in the testis. Therefore, TSPYL 1, 2 and 4 were included in the present study. To better understand how TSPYL1, 2, and 4 might influence CYP expression, we performed a series of pull-downs and mass spectrometric analyses. Panther pathway analysis of the 2272 pulled down proteins for all 3 TSPYL isoforms showed that the top five pathways were the Wnt signaling pathway, the Integrin signaling pathway, the Gonadotropin releasing hormone receptor pathway, the Angiogenesis pathway and Inflammation mediated by chemokines and cytokines. Specifically, we observed that 177 Wnt signaling pathway proteins were pulled down with the TSPYLs. Subsequent luciferase assays showed that TSPYL1 knockdown had a greater effect on the activation of Wnt signaling than did TSPYL2 or TSPYL4 knockdown. Therefore, in subsequent experiments, we focused our attention on TSPYL1. HepaRG cell qRT-PCR showed that TSPYL1 regulated the expression of CYPs involved in cholesterol-metabolism such as CYP1B1 and CYP7A1. Furthermore, TSPYL1 and β-catenin regulated CYP1B1 expression in opposite directions and TSPYL1 appeared to regulate CYP1B1 expression by blocking β-catenin binding to the TCF7L2 transcription factor on the CYP1B1 promoter. In β-catenin and TSPYL1 double knockdown cells, CYP1B1 expression and the generation of CYP1B1 downstream metabolites such as 20-HETE could be restored. Finally, we observed that TSPYL1 expression was associated with plasma cholesterol levels and BMI during previous clinical studies of obesity. In conclusion, this series of experiments has revealed a novel mechanism for regulation of the expression of cholesterol-metabolizing CYPs, particularly CYP1B1, by TSPYL1 via Wnt/β-catenin signaling, raising the possibility that TSPYL1 might represent a molecular target for influencing cholesterol homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
49
|
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
|
50
|
Wen P, Wang H, Li Y, Sui X, Hou Z, Guo X, Xue W, Liu D, Wang Y, Gao J. MICALL2 as a substrate of ubiquitinase TRIM21 regulates tumorigenesis of colorectal cancer. Cell Commun Signal 2022; 20:170. [DOI: 10.1186/s12964-022-00984-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/01/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Molecule interacting with CasL-like protein 2 (MICALL2) is believed to regulate cytoskeleton dynamics, tight junction formation, and neurite outgrowth. However, its biological role and the underlying mechanism in colorectal cancer (CRC) remain largely elusive.
Methods
qRT-PCR, Western blotting and immunohistochemistry assays were used to detect the expression levels of different genes. Next, mass spectrometry, co-immunoprecipitation and immunofluorescence staining were used to detect the interactions of proteins. Furthermore, MTT assay, colony formation assay, wound-healing assays and xenograft tumor models were performed to demonstrate the functions of MICALL2 in CRC. In addition, transcriptome sequencing and Western blotting were conducted to verify the mechanism of MICALL2 in CRC.
Results
We found that both mRNA and protein levels of MICALL2 are up-regulated in colorectal cancer tissues compared with non-tumor tissues and that its overexpression is closely correlated with poor prognosis. Ubiquitin E3 ligase Tripartite motif-containing protein 21 (TRIM21) mediated MICALL2 ubiquitination and proteasome-dependent degradation, negatively correlated with MICALL2 levels, and reversely regulated the tumorigenic activity of MICALL2 in CRC. Functional studies confirmed that MICALL2 promoted colorectal cancer cell growth and migration via the Wnt/β-catenin signaling pathway.
Conclusions
As a substrate of ubiquitinase TRIM21, MICALL2 enhances the growth and migration of colorectal cancer cells and activates the Wnt/β-catenin signaling pathway.
Collapse
|