1
|
Salem MM, Elkeiy MM, El-Gamal MM, Saad-Allah KM, Khamis AA. Evaluation of Paracentrotus lividus spines extract antioxidant, antidiabetic, anti-inflammatory, antimicrobial, and mechanistic anticancer: Insights into its composition using UPLC-ESI-MS-based metabolomic profiling. Tissue Cell 2025; 94:102808. [PMID: 40010182 DOI: 10.1016/j.tice.2025.102808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/01/2025] [Accepted: 02/17/2025] [Indexed: 02/28/2025]
Abstract
Sea urchins are widely distributed in waters worldwide. The present study carried out the in vitro metabolomic bioactivity profiling using UPLCMS/MS of Paracentrotus lividus spines extract. Investigations were also conducted on molecular anticancer, anti-inflammatory, antidiabetic, antioxidant, and antibacterial properties. A comprehensive untargeted metabolic profiling of P. lividus spines extract resulted in the classification of more than 13 metabolites. Their metabolomic quantitative evaluations were assumed by measuring total phenolic, flavonoids, dihydroflavonol, sugar, and protein contents. The P. lividus spines extract exhibited powerful antioxidant capacity using DPPH*, ABTS+, reducing power, and phosphomolybdate assays. Moreover, P. lividus spines extract highly elucidated antidiabetic and anti-inflammatory activity by inhibiting α-amylase enzyme and protein denaturation. Further, the spines of the P. lividus exhibited significant antibacterial effects. Besides, extract from P. lividus spines showed a strong cytotoxic impact against a variety of HepG-2 and MCF-7 cancer cell lines. It was discovered that the P. lividus spines extract triggered cell cycle arrest in the sub-G0/G1 phase and suppressed the growth of cancer cells via suppressing mRNA of Akt/MAPK/Bcl-2/c-myc and protein expression of β-Catenin/ki-67. Conclusively, the extract derived from the spines of the sea urchin species P. lividus demonstrates significant potential for utilization in various pharmaceutical industries.
Collapse
Affiliation(s)
- Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Mai M Elkeiy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Mona M El-Gamal
- Zoology Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Khalil M Saad-Allah
- Botany Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Abeer A Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
2
|
Khairi S, Wang CY, Anuraga G, Prayugo FB, Ansar M, Lesmana MHS, Irham LM, Shen CY, Chung MH. Integrative Analysis of DNA Methylation and microRNA Reveals GNPDA1 and SLC25A16 Related to Biopsychosocial Factors Among Taiwanese Women with a Family History of Breast Cancer. J Pers Med 2025; 15:134. [PMID: 40278313 PMCID: PMC12028518 DOI: 10.3390/jpm15040134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Biopsychosocial factors, including family history, influence the development of breast cancer. Malignancies in women with a family history of breast cancer may be detectable based on DNA methylation and microRNA. Objectives: The present study extended an integrative analysis of DNA methylation and microRNA to identify genes associated with biopsychosocial factors. Methods: We identified 3060 healthy women from the Taiwan Biobank and included 32 blood plasma samples for analysis of biopsychosocial factors and epigenetic changes. GEO databases and bioinformatics approaches were used for the identification and validation of potential genes. Results: Our integrative analysis revealed GNPDA1 and SLC25A16 as potential genes. Age, a family history of cancer, and alcohol consumption were associated with GNPDA1 and SLC25A16 based on the current data set and the GEO data set. GNPDA1 and SLC25A16 exhibited significant expression in breast cancer tissues based on UALCAN analysis, where they were overexpressed and underexpressed, respectively. Through a MethSurv analysis, GNPDA1 hypomethylation and SLC25A16 hypermethylation were associated with poor prognoses in terms of overall survival in breast cancer. Moreover, through a MetaCore functional enrichment analysis, GNPDA1 and SLC25A16 were associated with the BRCA1, BRCA2, and pro-oncogenic actions of the androgen receptor in breast cancer. Further, GNPDA1 and SLC25A16 were enriched in known targets of approved cancer drugs as potential genes associated with breast cancer. Conclusions: These two genes might serve as biomarkers for the early detection of breast cancer, especially for women with a family history of breast cancer.
Collapse
Affiliation(s)
- Sabiah Khairi
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
| | - Chih-Yang Wang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan;
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei City 11031, Taiwan
| | - Gangga Anuraga
- Department of Statistics, Faculty of Science and Technology, Universitas PGRI Adi Buana, Surabaya 60234, Indonesia;
| | - Fidelia Berenice Prayugo
- Chang Gung Medical Education Research Centre (CG-MERC), Chang Gung Memorial Hospital, Taoyuan City 33302, Taiwan;
- School of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Muhamad Ansar
- Ph.D. Program in the Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei City 110301, Taiwan;
| | - Mohammad Hendra Setia Lesmana
- Department of Mental Health and Community, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia;
| | | | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei City 11031, Taiwan
- College of Public Health, China Medical University, Taichung City 406040, Taiwan
| | - Min-Huey Chung
- School of Nursing, College of Nursing, Taipei Medical University, Taipei City 11031, Taiwan;
- Department of Nursing, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
| |
Collapse
|
3
|
Hassan D, Menges CW, Testa JR, Bellacosa A. AKT kinases as therapeutic targets. J Exp Clin Cancer Res 2024; 43:313. [PMID: 39614261 PMCID: PMC11606119 DOI: 10.1186/s13046-024-03207-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/03/2024] [Indexed: 12/01/2024] Open
Abstract
AKT, or protein kinase B, is a central node of the PI3K signaling pathway that is pivotal for a range of normal cellular physiologies that also underlie several pathological conditions, including inflammatory and autoimmune diseases, overgrowth syndromes, and neoplastic transformation. These pathologies, notably cancer, arise if either the activity of AKT or its positive or negative upstream or downstream regulators or effectors goes unchecked, superimposed on by its intersection with a slew of other pathways. Targeting the PI3K/AKT pathway is, therefore, a prudent countermeasure. AKT inhibitors have been tested in many clinical trials, primarily in combination with other drugs. While some have recently garnered attention for their favorable profile, concern over resistance and off-target effects have continued to hinder their widespread adoption in the clinic, mandating a discussion on alternative modes of targeting. In this review, we discuss isoform-centric targeting that may be more effective and less toxic than traditional pan-AKT inhibitors and its significance for disease prevention and treatment, including immunotherapy. We also touch on the emerging mutant- or allele-selective covalent allosteric AKT inhibitors (CAAIs), as well as indirect, novel AKT-targeting approaches, and end with a briefing on the ongoing quest for more reliable biomarkers predicting sensitivity and response to AKT inhibitors, and their current state of affairs.
Collapse
Affiliation(s)
- Dalal Hassan
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Thomas Jefferson University, 901 Walnut St, Philadelphia, PA, 19107, USA
| | - Craig W Menges
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Joseph R Testa
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Alfonso Bellacosa
- Nuclear Dynamics and Cancer Program, Cancer Epigenetics Institute, Institute for Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
| |
Collapse
|
4
|
Tao W, Xufeng Y, Xianmei C, Mengrou Q, Jieqiong W, Mingqi Q. Exploring the Mechanism of Myrrh in the Treatment of Breast Cancer Based on Network Pharmacology and Cell Experiments. Chem Biol Drug Des 2024; 104:e14604. [PMID: 39147995 DOI: 10.1111/cbdd.14604] [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: 08/17/2023] [Revised: 06/23/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
This study aimed to investigate the mechanism of action of myrrh in breast cancer (BC) treatment and identify its effective constituents. Data on the compounds and targets of myrrh were collected from the TCMSP, PubChem, and Swiss Target Prediction databases. BC-related targets were obtained from the Genecard database. A protein-protein interaction (PPI) analysis, gene ontology (GO) enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted on the intersecting targets of the disease and drug. The key targets of myrrh in BC treatment were identified based on the PPI network. The active constituents of myrrh were determined through reverse-screening using the top 20 KEGG pathways. Macromolecular docking studies, molecular dynamic (MD) simulations, and cell assays were utilized to validate the active constituents and critical targets. Network pharmacology indicated that VEGFA, TP53, ESR1, EGFR, and AKT1 are key targets of myrrh. Pelargonidin chloride, Quercetin, and Naringenin were identified as the active constituents of myrrh. Macromolecular docking showed that Quercetin and Naringenin have strong docking capabilities with ESR1. The results of MD simulation experiments align with those of molecular docking experiments. Cell and western blot assays demonstrated that Quercetin and Naringenin could inhibit MCF-7 cells and significantly reduce the expression of ESR1 protein. The findings reveal the active constituents, key targets, and molecular mechanisms of myrrh in BC treatment, providing scientific evidence that supports the role of myrrh in BC therapy. Furthermore, the results suggest that network pharmacology predictions require experimental validation for reliability.
Collapse
Affiliation(s)
- Wu Tao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Traditional Chinese Medicine, Research and Innovation Team of Emotional Diseases and Syndrome Research, Shandong University of Traditional Chinese Medicine, Jinan, China
- Emotional Disease Syndrome Innovative Chinese Medicine Research Young Scientific Research and Innovation Team, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Xufeng
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Traditional Chinese Medicine, Research and Innovation Team of Emotional Diseases and Syndrome Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chen Xianmei
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Traditional Chinese Medicine, Research and Innovation Team of Emotional Diseases and Syndrome Research, Shandong University of Traditional Chinese Medicine, Jinan, China
- Emotional Disease Syndrome Innovative Chinese Medicine Research Young Scientific Research and Innovation Team, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qu Mengrou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Traditional Chinese Medicine, Research and Innovation Team of Emotional Diseases and Syndrome Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wang Jieqiong
- Emotional Disease Syndrome Innovative Chinese Medicine Research Young Scientific Research and Innovation Team, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiao Mingqi
- College of Traditional Chinese Medicine, Research and Innovation Team of Emotional Diseases and Syndrome Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Lucotte EA, Asgari Y, Sugier PE, Karimi M, Domenighetti C, Lesueur F, Boland-Augé A, Ostroumova E, de Vathaire F, Zidane M, Guénel P, Deleuze JF, Boutron-Ruault MC, Severi G, Liquet B, Truong T. Investigation of common genetic risk factors between thyroid traits and breast cancer. Hum Mol Genet 2023; 33:38-47. [PMID: 37740403 PMCID: PMC10729861 DOI: 10.1093/hmg/ddad159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023] Open
Abstract
Breast cancer (BC) risk is suspected to be linked to thyroid disorders, however observational studies exploring the association between BC and thyroid disorders gave conflicting results. We proposed an alternative approach by investigating the shared genetic risk factors between BC and several thyroid traits. We report a positive genetic correlation between BC and thyroxine (FT4) levels (corr = 0.13, p-value = 2.0 × 10-4) and a negative genetic correlation between BC and thyroid-stimulating hormone (TSH) levels (corr = -0.09, p-value = 0.03). These associations are more striking when restricting the analysis to estrogen receptor-positive BC. Moreover, the polygenic risk scores (PRS) for FT4 and hyperthyroidism are positively associated to BC risk (OR = 1.07, 95%CI: 1.00-1.13, p-value = 2.8 × 10-2 and OR = 1.04, 95%CI: 1.00-1.08, p-value = 3.8 × 10-2, respectively), while the PRS for TSH is inversely associated to BC risk (OR = 0.93, 95%CI: 0.89-0.97, p-value = 2.0 × 10-3). Using the PLACO method, we detected 49 loci associated to both BC and thyroid traits (p-value < 5 × 10-8), in the vicinity of 130 genes. An additional colocalization and gene-set enrichment analyses showed a convincing causal role for a known pleiotropic locus at 2q35 and revealed an additional one at 8q22.1 associated to both BC and thyroid cancer. We also found two new pleiotropic loci at 14q32.33 and 17q21.31 that were associated to both TSH levels and BC risk. Enrichment analyses and evidence of regulatory signals also highlighted brain tissues and immune system as candidates for obtaining associations between BC and TSH levels. Overall, our study sheds light on the complex interplay between BC and thyroid traits and provides evidence of shared genetic risk between those conditions.
Collapse
Affiliation(s)
- Elise A Lucotte
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| | - Yazdan Asgari
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| | - Pierre-Emmanuel Sugier
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
- Laboratoire de Mathématiques et de leurs Applications de Pau, Université de Pau et des Pays de l’Adour, UMR CNRS 5142, E2S-UPPA, 64013 Pau, France
| | - Mojgan Karimi
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| | - Cloé Domenighetti
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| | - Fabienne Lesueur
- Inserm, U900, Institut Curie, PSL University, Mines ParisTech, 75006 Paris, France
| | - Anne Boland-Augé
- National Centre of Human Genomics Research, François Jacob Institute of Biology, Commissariat à l’Energie Atomique, Paris-Saclay University, 91000 Evry, France
| | | | - Florent de Vathaire
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team of Epidemiology of radiations, 94807 Villejuif, France
| | - Monia Zidane
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team of Epidemiology of radiations, 94807 Villejuif, France
| | - Pascal Guénel
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| | - Jean-François Deleuze
- National Centre of Human Genomics Research, François Jacob Institute of Biology, Commissariat à l’Energie Atomique, Paris-Saclay University, 91000 Evry, France
| | | | - Gianluca Severi
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
- Department of Statistics, Computer Science, Applications “G. Parenti”, University of Florence, 50121 Florence, Italy
| | - Benoît Liquet
- Laboratoire de Mathématiques et de leurs Applications de Pau, Université de Pau et des Pays de l’Adour, UMR CNRS 5142, E2S-UPPA, 64013 Pau, France
- School of Mathematical and Physical Sciences, Macquarie University, 2109 Sydney, Australia
| | - Thérèse Truong
- Paris-Saclay University, UVSQ, Gustave Roussy, Inserm, CESP, Team “Exposome and Heredity”, 94807 Villejuif, France
| |
Collapse
|
6
|
Wylaź M, Kaczmarska A, Pajor D, Hryniewicki M, Gil D, Dulińska-Litewka J. Exploring the role of PI3K/AKT/mTOR inhibitors in hormone-related cancers: A focus on breast and prostate cancer. Biomed Pharmacother 2023; 168:115676. [PMID: 37832401 DOI: 10.1016/j.biopha.2023.115676] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Breast cancer (BC) and prostate cancer (PC) are at the top of the list when it comes to the most common types of cancers worldwide. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway is important, in that it strongly influences the development and progression of these tumors. Previous studies have emphasized the key role of inhibitors of the PIK3/AKT/mTOR signaling pathway in the treatment of BC and PC, and it remains to be a crucial method of treatment. In this review, the inhibitors of these signaling pathways are compared, as well as their effectiveness in therapy and potential as therapeutic agents. The use of these inhibitors as polytherapy is evaluated, especially with the use of hormonal therapy, which has shown promising results.
Collapse
Affiliation(s)
- Mateusz Wylaź
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Anna Kaczmarska
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dawid Pajor
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Matthew Hryniewicki
- Student Scientific Group at Jagiellonian University Medical College, Faculty of Medicine, Medical Biochemistry, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Dorota Gil
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Mikołaja Kopernika Street 7C, 31-034 Krakow, Poland.
| |
Collapse
|
7
|
Vuorinen SI, Okolicsanyi RK, Gyimesi M, Meyjes-Brown J, Saini D, Pham SH, Griffiths LR, Haupt LM. SDC4-rs1981429 and ATM-rs228590 may provide early biomarkers of breast cancer risk. J Cancer Res Clin Oncol 2023; 149:4563-4578. [PMID: 36152082 PMCID: PMC10349731 DOI: 10.1007/s00432-022-04236-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/23/2022] [Indexed: 01/20/2023]
Abstract
In Australia, 13% of women are diagnosed with breast cancer (BC) in their lifetime with approximately 20,000 women diagnosed with the disease in 2021. BC is characterised by complex histological and genomic influences with recent advances in cancer biology improving early diagnosis and personalised treatment interventions. The Phosphatidyl-inositol-3-kinase/Protein kinase B (PI3K/AKT) pathway is essential in apoptosis resistance, cell survival, activation of cellular responses to DNA damage and DNA repair. Heparan sulfate proteoglycans (HSPGs) are ubiquitous molecules found on the cell surface and in the extracellular matrix with essential functions in regulating cell survival, growth, adhesion and as mediators of cell differentiation and migration. HSPGs, particularly the syndecans (SDCs), have been linked to cancers, making them an exciting target for anticancer treatments. In the PI3K/AKT pathway, syndecan-4 (SDC4) has been shown to downregulate AKT Serine/Threonine Kinase (AKT1) gene expression, while the ATM Serine/Threonine Kinase (ATM) gene has been found to inhibit this pathway upstream of AKT. We investigated single-nucleotide polymorphisms (SNPs) in HSPG and related genes SDC4, AKT1 and ATM and their influence on the prevalence of BC. SNPs were genotyped in the Australian Caucasian Genomics Research Centre Breast Cancer (GRC-BC) population and in the Griffith University-Cancer Council Queensland Breast Cancer Biobank (GU-CCQ BB) population. We identified that SDC4-rs1981429 and ATM-rs228590 may influence the development and progression of BC, having the potential to become biomarkers in early BC diagnosis and personalised treatment.
Collapse
Affiliation(s)
- Sofia I Vuorinen
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Martina Gyimesi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Jacob Meyjes-Brown
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Deepa Saini
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Son H Pham
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Lyn R Griffiths
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave, Kelvin Grove, QLD, 4059, Australia.
| |
Collapse
|
8
|
Saha ST, Abdulla N, Zininga T, Shonhai A, Wadee R, Kaur M. 2-Hydroxypropyl-β-cyclodextrin (HPβCD) as a Potential Therapeutic Agent for Breast Cancer. Cancers (Basel) 2023; 15:2828. [PMID: 37345165 PMCID: PMC10216648 DOI: 10.3390/cancers15102828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Cholesterol accumulation is documented in various malignancies including breast cancer. Consequently, depleting cholesterol in cancer cells can serve as a viable treatment strategy. We identified the potency of 2-hydroxypropyl-β-cyclodextrin (HPβCD), a cholesterol-depletor in vitro against two breast cancer cell lines: MCF-7 (Oestrogen-receptor positive, ER+) and MDA-MB-231 (Triple negative breast cancer (TNBC)). The results were then compared against two non-cancerous cell lines using cytotoxic-, apoptosis-, and cholesterol-based assays. Treatment with HPβCD showed preferential and significant cytotoxic potential in cancer cells, inducing apoptosis in both cancer cell lines (p < 0.001). This was mediated due to significant depletion of cholesterol (p < 0.001). We further tested HPβCD in a MF-1 mice (n = 14) xenograft model and obtained 73.9%, 94% and 100% reduction in tumour size for late-, intermediate-, and early-stage TNBC, respectively. We also detected molecular-level perturbations in the expression patterns of several genes linked to breast cancer and cholesterol signalling pathways using RT2-PCR arrays and have identified SFRP1 as a direct binding partner to HPβCD through SPR drug interaction analysis. This work unravels mechanistic insights into HPβCD-induced cholesterol depletion, which leads to intrinsic apoptosis induction. Results from this study potentiate employing cholesterol depletion as a promising unconventional anticancer therapeutic strategy, which warrants future clinical investigations.
Collapse
Affiliation(s)
- Sourav Taru Saha
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS-2050, Johannesburg 2050, South Africa
| | - Naaziyah Abdulla
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS-2050, Johannesburg 2050, South Africa
| | - Tawanda Zininga
- Department of Biochemistry and Microbiology, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa
- Department of Biochemistry, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Addmore Shonhai
- Department of Biochemistry and Microbiology, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa
| | - Reubina Wadee
- Department of Anatomical Pathology, School of Pathology, University of the Witwatersrand/National Health Laboratory Service, Johannesburg 2000, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS-2050, Johannesburg 2050, South Africa
| |
Collapse
|
9
|
Li M, Lin C, Cai Z. Breast cancer stem cell-derived extracellular vesicles transfer ARRDC1-AS1 to promote breast carcinogenesis via a miR-4731-5p/AKT1 axis-dependent mechanism. Transl Oncol 2023; 31:101639. [PMID: 36801666 PMCID: PMC9971553 DOI: 10.1016/j.tranon.2023.101639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/17/2023] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
OBJECTIVES Deregulation of long non-coding RNAs (lncRNAs) has been frequently reported in breast cancer (BC). This goes to show the importance of understanding its significant contribution towards breast carcinogenesis. In the present study, we clarified a carcinogenic mechanism based on the ARRDC1-AS1 delivered by breast cancer stem cells-derived extracellular vesicles (BCSCs-EVs) in BC. METHODS The isolated and well characterized BCSCs-EVs were co-cultured with BC cells. The expression of ARRDC1-AS1, miR-4731-5p, and AKT1 was determined in BC cell lines. BC cells were assayed for their viability, invasion, migration and apoptosis in vitro by CCK-8, Transwell and flow cytometry, as well as tumor growth in vivo after loss- and gain-of function assays. Dual-luciferase reporter gene, RIP and RNA pull-down assays were performed to determine the interactions among ARRDC1-AS1, miR-4731-5p, and AKT1. RESULTS Elevation of ARRDC1-AS1 and AKT1 as well as miR-4731-5p downregulation were observed in BC cells. ARRDC1-AS1 was enriched in BCSCs-EVs. Furthermore, EVs containing ARRDC1-AS1 enhanced the BC cell viability, invasion and migration and glutamate concentration. Mechanistically, ARRDC1-AS1 elevated the expression of AKT1 by competitively binding to miR-4731-5p. ARRDC1-AS1-containing EVs were also found to enhance tumor growth in vivo. CONCLUSION Collectively, BCSCs-EVs-mediated delivery of ARRDC1-AS1 may promote the malignant phenotypes of BC cells via the miR-4731-5p/AKT1 axis.
Collapse
Affiliation(s)
- Mingzhu Li
- Area N4 of Surgical Oncology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 1028, Anji South Road, Fengze District, Quanzhou, Fujian 362000, China.
| | - Conglin Lin
- Area N4 of Surgical Oncology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 1028, Anji South Road, Fengze District, Quanzhou, Fujian 362000, China
| | - Zhibing Cai
- Area N4 of Surgical Oncology, Quanzhou First Hospital Affiliated to Fujian Medical University, No. 1028, Anji South Road, Fengze District, Quanzhou, Fujian 362000, China
| |
Collapse
|
10
|
Zhang J, Lin X, Chen Y, Li T, Lee AC, Chow EY, Cho WC, Chan T. LAFITE Reveals the Complexity of Transcript Isoforms in Subcellular Fractions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203480. [PMID: 36461702 PMCID: PMC9875686 DOI: 10.1002/advs.202203480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/28/2022] [Indexed: 06/17/2023]
Abstract
Characterization of the subcellular distribution of RNA is essential for understanding the molecular basis of biological processes. Here, the subcellular nanopore direct RNA-sequencing (DRS) of four lung cancer cell lines (A549, H1975, H358, and HCC4006) is performed, coupled with a computational pipeline, Low-abundance Aware Full-length Isoform clusTEr (LAFITE), to comprehensively analyze the full-length cytoplasmic and nuclear transcriptome. Using additional DRS and orthogonal data sets, it is shown that LAFITE outperforms current methods for detecting full-length transcripts, particularly for low-abundance isoforms that are usually overlooked due to poor read coverage. Experimental validation of six novel isoforms exclusively identified by LAFITE further confirms the reliability of this pipeline. By applying LAFITE to subcellular DRS data, the complexity of the nuclear transcriptome is revealed in terms of isoform diversity, 3'-UTR usage, m6A modification patterns, and intron retention. Overall, LAFITE provides enhanced full-length isoform identification and enables a high-resolution view of the RNA landscape at the isoform level.
Collapse
Affiliation(s)
- Jizhou Zhang
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
- State Key Laboratory of AgrobiotechnologyThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Xiao Lin
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
- State Key Laboratory of AgrobiotechnologyThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Yuelong Chen
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Tsz‐Ho Li
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
- State Key Laboratory of AgrobiotechnologyThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Alan Chun‐Kit Lee
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
| | | | | | - Ting‐Fung Chan
- School of Life SciencesThe Chinese University of Hong KongShatinHong Kong SARChina
- State Key Laboratory of AgrobiotechnologyThe Chinese University of Hong KongShatinHong Kong SARChina
| |
Collapse
|
11
|
Leal-Orta E, Ramirez-Ricardo J, Garcia-Hernandez A, Cortes-Reynosa P, Salazar EP. Extracellular vesicles from MDA-MB-231 breast cancer cells stimulated with insulin-like growth factor 1 mediate an epithelial-mesenchymal transition process in MCF10A mammary epithelial cells. J Cell Commun Signal 2022; 16:531-546. [PMID: 34309795 PMCID: PMC9733745 DOI: 10.1007/s12079-021-00638-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays an important role in function and development of the mammary gland. However, high levels of IGF-1 has been associated with an increased risk of breast cancer development. Epithelial-mesenchymal transition (EMT) is a process where epithelial cells lose their epithelial characteristics and acquire a mesenchymal phenotype, which is considered one of the most important mechanisms in cancer initiation and promotion of metastasis. Extracellular vesicles (EVs) are released into the extracellular space by different cell types, which mediate intercellular communication and play an important role in different physiological and pathological processes, such as cancer. In this study, we demonstrate that EVs from MDA-MB-231 breast cancer cells stimulated with IGF-1 (IGF-1 EVs) decrease the levels of E-cadherin, increase the expression of vimentin and N-cadherin and stimulate the secretion of metalloproteinase-9 in mammary non-tumorigenic epithelial cells MCF10A. IGF-1 EVs also induce the expression of Snail1, Twist1 and Sip1, which are transcription factors involved in EMT. Moreover, IGF-1 EVs induce activation of ERK1/2, Akt1 and Akt2, migration and invasion. In summary, we demonstrate, for the first time, that IGF-1 EVs induce an EMT process in mammary non-tumorigenic epithelial cells MCF10A.
Collapse
Affiliation(s)
- Elizabeth Leal-Orta
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| | | | | | - Pedro Cortes-Reynosa
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| | - Eduardo Perez Salazar
- grid.512574.0Departamento de Biologia Celular, Cinvestav-IPN, 07360 Mexico City, Mexico
| |
Collapse
|
12
|
Song Y, Wang J, Wang X, Zhang H, Niu X, Yang Y, Yang X, Yin L, Wang Y, Zhang C, Shui R, Zhang Q, Ji H. Analyzing the multi-target pharmacological mechanism of folium Artemisia argyi acting on breast cancer: a network pharmacology approach. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1368. [PMID: 36660662 PMCID: PMC9843367 DOI: 10.21037/atm-22-5769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Background Folium Artemisia argyi (FAA) is a traditional Chinese herbal medicine that is widely used in the clinic. However, the underlying mechanisms of its anticancer effects have not been fully elucidated. Methods In this study, we applied a network pharmacology approach to identify the potential mechanisms of FAA against breast cancer. To be specific, we screened the active ingredients and potential targets of the FAA through the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. Meanwhile, we employed the oral bioavailability (OB) and drug-likeness (DL) to search for potential bioactive compounds of FAA. Breast cancer-related target genes data were gathered from the GeneCards and Online Mendelian Inheritance in Man (OMIM) databases, and the protein-protein interaction (PPI) data were acquired from the Search Tool for the Retrieval of Interacting Genes (STRING) database. In addition, we constructed the network and performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Enrichment Analysis. Results We obtained a total of nine active ingredients and 236 potential targets from FAA to construct a network, which showed that quercetin served as the major ingredient in FAA. AKT1 (RAC-alpha serine/threonine-protein kinase), MYC (Myc proto-oncogene protein), CASP3 (Caspase-3), EGFR (Epidermal growth factor receptor), JUN (Transcription factor AP-1), CCND1 (G1/S-specific cyclin-D1), VEGFA (Vascular endothelial growth factor A), ESR1 (Estrogen receptor), MAPK1 (Mitogen-activated protein kinase 1), and EGF (pro-epidermal growth factor) were identified as key targets of FAA in the treatment of breast cancer. The PPI cluster demonstrated that AKT1 was the seed in this cluster, indicating that AKT1 played a crucial role in connecting other nodes in the PPI network. This enrichment demonstrated that FAA was highly related to signal transduction, endocrine system, replication and repair, as well as cell growth and death. The enrichment results also verified that the underlying mechanisms of FAA against breast cancer might be attributed to the coordinated regulation of several cancer-related pathways, such as the MAPK and mammalian target of rapamycin (mTOR) signaling pathways, among others. Conclusions This study identified the potential targets and pathways of FAA in the treatment of breast cancer using a network pharmacology approach, and systematically elucidated the mechanisms of FAA in the treatment of breast cancer.
Collapse
Affiliation(s)
- Ying Song
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Jinlu Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xiuli Wang
- Department of Clinical Laboratory, The Seventh Hospital in Qiqihar, Qiqihar, China
| | - Han Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xingjian Niu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yue Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xudong Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Lei Yin
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yiran Wang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Cuiying Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Ruixue Shui
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
13
|
The Mechanism of Ginseng and Astragalus Decoction in the Treatment of Malignant Pleural Effusion Based on Network Pharmacology and Molecular Docking Technology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7731402. [PMID: 35341150 PMCID: PMC8941536 DOI: 10.1155/2022/7731402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/31/2022] [Indexed: 11/17/2022]
Abstract
Introduction The objective of our study is to explore the potential active ingredients and activity of Ginseng and Astragalus decoction (GAD) in the treatment of malignant pleural effusion (MPE) by using network pharmacology and molecular docking technologies. Methods The active ingredients and corresponding targets of Ginseng and Astragalus were extracted from the Traditional Chinese Medicine System Pharmacology Database and Analysis Platform. The relevant targets of malignant pleural effusion (MPE) were searched in the disease databases. Overlapping targets of Ginseng and Astragalus and the corresponding targets of MPE were obtained to define the effective target of GAD for the treatment of MPE. The STRING database was applied to construct a predicted protein-protein interaction network for intersected targets. The Cytoscape software was used to screen key targets with a therapeutic potential. Using the Metascape database, we performed Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analysis on the targets identified in the study. PyMOL and AutoDock Vina were used to molecularly dock the selected key components to their respective key targets for MPE treatment. Results The core target network revealed 22 main active ingredients, 26 main targets, and 16 signaling pathways in GAD. Molecular docking revealed 6 targets (AKT serine/threonine kinase 1, intercellular adhesion molecule, Jun proto-oncogene, peroxisome proliferator activated receptor gamma, prostaglandin-endoperoxide synthase 2, and tumor necrosis factor) that could partially dock with kaempferol, frutinone A, ginsenoside RH2, formononetin, and quercetin. Conclusions Several components, targets, and signaling pathways of GAD contribute to the treatment of MPE, which suggests a rationale for further investigation on GAD's active molecule and mechanism of action in the clinical application of MPE.
Collapse
|
14
|
Tatli O, Dinler Doganay G. Recent Developments in Targeting RAS Downstream Effectors for RAS-Driven Cancer Therapy. Molecules 2021; 26:molecules26247561. [PMID: 34946644 PMCID: PMC8703923 DOI: 10.3390/molecules26247561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Aberrant activity of oncogenic rat sarcoma virus (RAS) protein promotes tumor growth and progression. RAS-driven cancers comprise more than 30% of all human cancers and are refractory to frontline treatment strategies. Since direct targeting of RAS has proven challenging, efforts have been centered on the exploration of inhibitors for RAS downstream effector kinases. Two major RAS downstream signaling pathways, including the Raf/MEK/Erk cascade and the phosphatidylinositol-3-kinase (PI3K) pathway, have become compelling targets for RAS-driven cancer therapy. However, the main drawback in the blockade of a single RAS effector is the multiple levels of crosstalk and compensatory mechanisms between these two pathways that contribute to drug resistance against monotherapies. A growing body of evidence reveals that the sequential or synergistic inhibition of multiple RAS effectors is a more convenient route for the efficacy of cancer therapy. Herein, we revisit the recent developments and discuss the most promising modalities targeting canonical RAS downstream effectors for the treatment of RAS-driven cancers.
Collapse
Affiliation(s)
- Ozge Tatli
- Department of Molecular Biology, Genetics-Biotechnology, Graduate School, Istanbul Technical University, Istanbul 34469, Turkey;
- Department of Molecular Biology and Genetics, Istanbul Medeniyet University, Istanbul 34720, Turkey
| | - Gizem Dinler Doganay
- Department of Molecular Biology, Genetics-Biotechnology, Graduate School, Istanbul Technical University, Istanbul 34469, Turkey;
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul 34469, Turkey
- Correspondence: ; Tel.: +90-2122-857-256
| |
Collapse
|
15
|
Wu Z, Sun S, Fan R, Wang Z. tubulin alpha 1c promotes aerobic glycolysis and cell growth through upregulation of yes association protein expression in breast cancer. Anticancer Drugs 2021; 33:132-141. [PMID: 34845165 DOI: 10.1097/cad.0000000000001250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Tubulin alpha 1c (TUBA1C) as a member of α-tubulin was identified to take part in the occurrence and development of hepatocellular carcinoma and pancreatic cancer. Using the bioinformatics, we noticed that TUBA1C level was also increased in breast cancer was also demonstrated. Here, we explored TUBA1 role in modulation of breast cancer cell aerobic glycolysis, growth and migration and explored whether yes association protein (YAP) was involved. Fifty-five matched breast cancer tissues and the para-carcinoma normal tissues were included in this study and used to verify TUBA1C expression using quantitative reverse transcription-PCR and western blotting. ATP level, lactate secretion and glucose consumption were used to assess aerobic glycolysis. Cell growth, invasion, migration and tumorigenesis were detected using cell count kit-8, transwell, wound healing and animal assays. TUBA1 was upregulated in breast cancer, which associated with advanced primary tumor, lymph node, metastasis stage and tumor size. Silencing of TUBA1C with sh-TUBA1C infection led to significant inhibitions in ATP level, lactate secretion, glucose consumption, cell growth, migration, invasion and tumorigenesis, as well as declined YAP expression, while TUBA1C overexpression induced a opposite result. And, the above tendencies induced by TUBA1C downregulation were reversed by YAP overexpression. This study revealed that TUBA1C was overexpressed in breast cancer and promoted aerobic glycolysis and cell growth through upregulation of YAP expression.
Collapse
Affiliation(s)
- Zhu Wu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan Department of respiratory medicine, Jingzhou first people's Hospital Department of breast surgery, Jingzhou first people's Hospital, Jingzhou, Hubei, China
| | | | | | | |
Collapse
|
16
|
Akt Isoforms: A Family Affair in Breast Cancer. Cancers (Basel) 2021; 13:cancers13143445. [PMID: 34298660 PMCID: PMC8306188 DOI: 10.3390/cancers13143445] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Breast cancer is the second leading cause of cancer-related death in women in the United States. The Akt signaling pathway is deregulated in approximately 70% of patients with breast cancer. While targeting Akt is an effective therapeutic strategy for the treatment of breast cancer, there are several members in the Akt family that play distinct roles in breast cancer. However, the function of Akt isoforms depends on many factors. This review analyzes current progress on the isoform-specific functions of Akt isoforms in breast cancer. Abstract Akt, also known as protein kinase B (PKB), belongs to the AGC family of protein kinases. It acts downstream of the phosphatidylinositol 3-kinase (PI3K) and regulates diverse cellular processes, including cell proliferation, cell survival, metabolism, tumor growth and metastasis. The PI3K/Akt signaling pathway is frequently deregulated in breast cancer and plays an important role in the development and progression of breast cancer. There are three closely related members in the Akt family, namely Akt1(PKBα), Akt2(PKBβ) and Akt3(PKBγ). Although Akt isoforms share similar structures, they exhibit redundant, distinct as well as opposite functions. While the Akt signaling pathway is an important target for cancer therapy, an understanding of the isoform-specific function of Akt is critical to effectively target this pathway. However, our perception regarding how Akt isoforms contribute to the genesis and progression of breast cancer changes as we gain new knowledge. The purpose of this review article is to analyze current literatures on distinct functions of Akt isoforms in breast cancer.
Collapse
|
17
|
Zhao Y, Fu X, Lopez JI, Rowan A, Au L, Fendler A, Hazell S, Xu H, Horswell S, Shepherd STC, Spain L, Byrne F, Stamp G, O'Brien T, Nicol D, Augustine M, Chandra A, Rudman S, Toncheva A, Pickering L, Sahai E, Larkin J, Bates PA, Swanton C, Turajlic S, Litchfield K. Selection of metastasis competent subclones in the tumour interior. Nat Ecol Evol 2021; 5:1033-1045. [PMID: 34002049 PMCID: PMC7611703 DOI: 10.1038/s41559-021-01456-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
The genetic evolutionary features of solid tumour growth are becoming increasingly well described, but the spatial and physical nature of subclonal growth remains unclear. Here, we utilize 102 macroscopic whole-tumour images from clear cell renal cell carcinoma patients, with matched genetic and phenotypic data from 756 biopsies. Utilizing a digital image processing pipeline, a renal pathologist marked the boundaries between tumour and normal tissue and extracted positions of boundary line and biopsy regions to X and Y coordinates. We then integrated coordinates with genomic data to map exact spatial subclone locations, revealing how genetically distinct subclones grow and evolve spatially. We observed a phenotype of advanced and more aggressive subclonal growth in the tumour centre, characterized by an elevated burden of somatic copy number alterations and higher necrosis, proliferation rate and Fuhrman grade. Moreover, we found that metastasizing subclones preferentially originate from the tumour centre. Collectively, these observations suggest a model of accelerated evolution in the tumour interior, with harsh hypoxic environmental conditions leading to a greater opportunity for driver somatic copy number alterations to arise and expand due to selective advantage. Tumour subclone growth is predominantly spatially contiguous in nature. We found only two cases of subclone dispersal, one of which was associated with metastasis. The largest subclones spatially were dominated by driver somatic copy number alterations, suggesting that a large selective advantage can be conferred to subclones upon acquisition of these alterations. In conclusion, spatial dynamics is strongly associated with genomic alterations and plays an important role in tumour evolution.
Collapse
Affiliation(s)
- Yue Zhao
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao Fu
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK
| | - Jose I Lopez
- Department of Pathology, Cruces University Hospital, Biocruces-Bizkaia Institute, Barakaldo, Spain
| | - Andrew Rowan
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Lewis Au
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK
| | - Annika Fendler
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
| | - Steve Hazell
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Hang Xu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart Horswell
- Department of Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK
| | - Scott T C Shepherd
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK
| | - Lavinia Spain
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK
| | - Fiona Byrne
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK
| | - Gordon Stamp
- Experimental Histopathology Laboratory, The Francis Crick Institute, London, UK
| | - Tim O'Brien
- Urology Centre, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - David Nicol
- Department of Urology, The Royal Marsden NHS Foundation Trust, London, UK
| | - Marcellus Augustine
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Ashish Chandra
- Department of Pathology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sarah Rudman
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Lisa Pickering
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK
| | - Erik Sahai
- Tumour Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - James Larkin
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK
| | - Paul A Bates
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK.
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Department of Medical Oncology, University College London Hospitals, London, UK.
| | - Samra Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London, UK.
- Renal and Skin Unit, the Royal Marsden NHS Foundation Trust, London, UK.
| | - Kevin Litchfield
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Tumour Immunogenomics and Immunosurveillance Laboratory, University College London Cancer Institute, London, UK.
| |
Collapse
|
18
|
Hinz N, Jücker M. AKT in Bone Metastasis of Solid Tumors: A Comprehensive Review. Cancers (Basel) 2021; 13:cancers13102287. [PMID: 34064589 PMCID: PMC8151478 DOI: 10.3390/cancers13102287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastasis is a frequent complication of solid tumors and leads to a reduced overall survival. Although much progress has been made in the field of tumor therapy in the last years, bone metastasis depicts a stage of the disease with a lack of appropriate therapeutical options. Hence, this review aims to present the role of AKT in bone metastasis of solid tumors to place the spotlight on AKT as a possible therapeutical approach for patients with bone metastases. Furthermore, we intended to discuss postulated underlying molecular mechanisms of the bone metastasis-promoting effect of AKT, especially in highly bone-metastatic breast, prostate, and lung cancer. To conclude, this review identified the AKT kinase as a potential therapeutical target in bone metastasis and revealed remaining questions, which need to be addressed in further research projects. Abstract Solid tumors, such as breast cancer and prostate cancer, often form bone metastases in the course of the disease. Patients with bone metastases frequently develop complications, such as pathological fractures or hypercalcemia and exhibit a reduced life expectancy. Thus, it is of vital importance to improve the treatment of bone metastases. A possible approach is to target signaling pathways, such as the PI3K/AKT pathway, which is frequently dysregulated in solid tumors. Therefore, we sought to review the role of the serine/threonine kinase AKT in bone metastasis. In general, activation of AKT signaling was shown to be associated with the formation of bone metastases from solid tumors. More precisely, AKT gets activated in tumor cells by a plethora of bone-derived growth factors and cytokines. Subsequently, AKT promotes the bone-metastatic capacities of tumor cells through distinct signaling pathways and secretion of bone cell-stimulating factors. Within the crosstalk between tumor and bone cells, also known as the vicious cycle, the stimulation of osteoblasts and osteoclasts also causes activation of AKT in these cells. As a consequence, bone metastasis is reduced after experimental inhibition of AKT. In summary, AKT signaling could be a promising therapeutical approach for patients with bone metastases of solid tumors.
Collapse
|
19
|
Degan SE, Gelman IH. Emerging Roles for AKT Isoform Preference in Cancer Progression Pathways. Mol Cancer Res 2021; 19:1251-1257. [PMID: 33931488 DOI: 10.1158/1541-7786.mcr-20-1066] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/01/2021] [Accepted: 04/27/2021] [Indexed: 12/16/2022]
Abstract
The phosphoinositol-3 kinase (PI3K)-AKT pathway is one of the most mutated in human cancers, predominantly associated with the loss of the signaling antagonist, PTEN, and to lesser extents, with gain-of-function mutations in PIK3CA (encoding PI3K-p110α) and AKT1. In addition, most oncogenic driver pathways activate PI3K/AKT signaling. Nonetheless, drugs targeting PI3K or AKT have fared poorly against solid tumors in clinical trials as monotherapies, yet some have shown efficacy when combined with inhibitors of other oncogenic drivers, such as receptor tyrosine kinases or nuclear hormone receptors. There is growing evidence that AKT isoforms, AKT1, AKT2, and AKT3, have different, often distinct roles in either promoting or suppressing specific parameters of oncogenic progression, yet few if any isoform-preferred substrates have been characterized. This review will describe recent data showing that the differential activation of AKT isoforms is mediated by complex interplays between PTEN, PI3K isoforms and upstream tyrosine kinases, and that the efficacy of PI3K/AKT inhibitors will likely depend on the successful targeting of specific AKT isoforms and their preferred pathways.
Collapse
Affiliation(s)
- Seamus E Degan
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| |
Collapse
|
20
|
You KS, Yi YW, Cho J, Seong YS. Dual Inhibition of AKT and MEK Pathways Potentiates the Anti-Cancer Effect of Gefitinib in Triple-Negative Breast Cancer Cells. Cancers (Basel) 2021; 13:1205. [PMID: 33801977 PMCID: PMC8000364 DOI: 10.3390/cancers13061205] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/28/2021] [Accepted: 03/07/2021] [Indexed: 12/14/2022] Open
Abstract
There is an unmet medical need for the development of new targeted therapeutic strategies for triple-negative breast cancer (TNBC). With drug combination screenings, we found that the triple combination of the protein kinase inhibitors (PKIs) of the epidermal growth factor receptor (EGFR), v-akt murine thymoma viral oncogene homolog (AKT), and MAPK/ERK kinase (MEK) is effective in inducing apoptosis in TNBC cells. A set of PKIs were first screened in combination with gefitinib in the TNBC cell line, MDA-MB-231. The AKT inhibitor, AT7867, was identified and further analyzed in two mesenchymal stem-like (MSL) subtype TNBC cells, MDA-MB-231 and HS578T. A combination of gefitinib and AT7867 reduced the proliferation and long-term survival of MSL TNBC cells. However, gefitinib and AT7867 induced the activation of the rat sarcoma (RAS)/ v-raf-1 murine leukemia viral oncogene homolog (RAF)/MEK/ extracellular signal-regulated kinase (ERK) pathway. To inhibit this pathway, MEK/ERK inhibitors were further screened in MDA-MB-231 cells in the presence of gefitinib and AT7867. As a result, we identified that the MEK inhibitor, PD-0325901, further enhanced the anti-proliferative and anti-clonogenic effects of gefitinib and AT7867 by inducing apoptosis. Our results suggest that the dual inhibition of the AKT and MEK pathways is a novel potential therapeutic strategy for targeting EGFR in TNBC cells.
Collapse
Affiliation(s)
- Kyu Sic You
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Korea;
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Korea;
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Korea;
| | - Yeon-Sun Seong
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Korea;
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Korea;
| |
Collapse
|
21
|
Hinz N, Baranowsky A, Horn M, Kriegs M, Sibbertsen F, Smit DJ, Clezardin P, Lange T, Schinke T, Jücker M. Knockdown of AKT3 Activates HER2 and DDR Kinases in Bone-Seeking Breast Cancer Cells, Promotes Metastasis In Vivo and Attenuates the TGFβ/CTGF Axis. Cells 2021; 10:cells10020430. [PMID: 33670586 PMCID: PMC7922044 DOI: 10.3390/cells10020430] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bone metastases frequently occur in breast cancer patients and lack appropriate treatment options. Hence, understanding the molecular mechanisms involved in the multistep process of breast cancer bone metastasis and tumor-induced osteolysis is of paramount interest. The serine/threonine kinase AKT plays a crucial role in breast cancer bone metastasis but the effect of individual AKT isoforms remains unclear. Therefore, AKT isoform-specific knockdowns were generated on the bone-seeking MDA-MB-231 BO subline and the effect on proliferation, migration, invasion, and chemotaxis was analyzed by live-cell imaging. Kinome profiling and Western blot analysis of the TGFβ/CTGF axis were conducted and metastasis was evaluated by intracardiac inoculation of tumor cells into NOD scid gamma (NSG) mice. MDA-MB-231 BO cells exhibited an elevated AKT3 kinase activity in vitro and responded to combined treatment with AKT- and mTOR-inhibitors. Knockdown of AKT3 significantly increased migration, invasion, and chemotaxis in vitro and metastasis to bone but did not significantly enhance osteolysis. Furthermore, knockdown of AKT3 increased the activity and phosphorylation of pro-metastatic HER2 and DDR1/2 but lowered protein levels of CTGF after TGFβ-stimulation, an axis involved in tumor-induced osteolysis. We demonstrated that AKT3 plays a crucial role in bone-seeking breast cancer cells by promoting metastatic potential without facilitating tumor-induced osteolysis.
Collapse
Affiliation(s)
- Nico Hinz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Anke Baranowsky
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Horn
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Malte Kriegs
- Department of Radiotherapy & Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- UCCH Kinomics Core Facility, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Freya Sibbertsen
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Daniel J. Smit
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Philippe Clezardin
- INSERM, Research Unit UMR S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, 69372 Lyon, France;
| | - Tobias Lange
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Thorsten Schinke
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
- Correspondence: ; Tel.: +49-(0)-40-7410-56339
| |
Collapse
|
22
|
Liang C, Niu J, Wang X, Zhang ZS, Yang RH, Yao X, Liu FY, Li WQ, Pei SH, Sun H, Wang CJ, Fang D, Xie SQ. P300-dependent acetylation of histone H3 is required for epidermal growth factor receptor-mediated high-mobility group protein A2 transcription in hepatocellular carcinoma. Cancer Sci 2020; 112:679-690. [PMID: 33164305 PMCID: PMC7894021 DOI: 10.1111/cas.14729] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/05/2020] [Accepted: 11/07/2020] [Indexed: 12/24/2022] Open
Abstract
High‐mobility group protein A2 (HMGA2) is highly expressed in hepatocellular carcinoma (HCC) cells and contributes to tumor metastasis and poor patient survival. However, the molecular mechanism through which HMGA2 is transcriptionally regulated in HCC cells remains largely unclear. Here, we showed that the expression HMGA2 was upregulated in HCC, and that elevated HMGA2 could promote tumor metastasis. Incubation of HCC cells with epidermal growth factor (EGF) could promote the expression of HMGA2 mRNA and protein. Mechanistic studies suggested that EGF can phosphorylate p300 at Ser1834 residue through the PI3K/Akt signaling pathway in HCC cells. Knockdown of p300 can reverse EGF‐induced HMGA2 expression and histone H3‐K9 acetylation, whereas a phosphorylation‐mimic p300 S1834D mutant can stimulate HMGA2 expression as well as H3‐K9 acetylation in HCC cells. Furthermore, we identified that p300‐mediated H3‐K9 acetylation participates in EGF‐induced HMGA2 expression in HCC. In addition, the levels of H3‐K9 acetylation positively correlated with the expression levels of HMGA2 in a chemically induced HCC model in rats and human HCC specimens.
Collapse
Affiliation(s)
- Chao Liang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Jie Niu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Xiao Wang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Zhan-Sheng Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Ruo-Han Yang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Xin Yao
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Fan-Ye Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Wen-Qi Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Shu-Hua Pei
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Hua Sun
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Chao-Jie Wang
- The Key Laboratory of Natural Medicine and Immuno-Engineering, Henan University, Kaifeng, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Song-Qiang Xie
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China.,Institute of Chemical Biology, School of Pharmacy, Henan University, Kaifeng, China
| |
Collapse
|
23
|
Wang X, Liang C, Yao X, Yang RH, Zhang ZS, Liu FY, Li WQ, Pei SH, Ma J, Xie SQ, Fang D. PKM2-Induced the Phosphorylation of Histone H3 Contributes to EGF-Mediated PD-L1 Transcription in HCC. Front Pharmacol 2020; 11:577108. [PMID: 33324209 PMCID: PMC7725877 DOI: 10.3389/fphar.2020.577108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 10/19/2020] [Indexed: 12/19/2022] Open
Abstract
High expression of programmed death-ligand-1 (PD-L1) in hepatocellular carcinoma (HCC) cells usually inhibits the proliferation and functions of T cells, leading to immune suppression in tumor microenvironment. However, very little has been described regarding the mechanism of PD-L1 overexpression in HCC cells. In the present study, we found epidermal growth factor (EGF) stimulation promoted the expression of PD-L1 mRNA and protein in HCC cells. Inhibition of epidermal growth factor receptor (EGFR) could reverse EGF-induced the expression of PD-L1 mRNA and protein. Subsequently, we also observed that the phosphorylation level of Pyruvate kinase isoform M2 (PKM2) at Ser37 site was also increased in response to EGF stimulation. Expression of a phosphorylation-mimic PKM2 S37D mutant stimulated PD-L1 expression as well as H3-Thr11 phosphorylation in HCC cells, while inhibition of PKM2 significantly blocked EGF-induced PD-L1 expression and H3-Thr11 phosphorylation. Furthermore, mutation of Thr11 of histone H3 into alanine abrogated EGF-induced mRNA and protein expression of PD-L1, Chromatin immunoprecipitation (ChIP) assay also suggested that EGF treatment resulted in enhanced H3-Thr11 phosphorylation at the PD-L1 promoter. In a diethylnitrosamine (DEN)-induced rat model of HCC, we found that the expression of phosphorylated EGFR, PKM2 nuclear expression, H3-Thr11 phosphorylation as well as PD-L1 mRNA and protein was higher in the livers than that in normal rat livers. Taken together, our study suggested that PKM2-dependent histone H3-Thr11 phosphorylation was crucial for EGF-induced PD-L1 expression at transcriptional level in HCC. These findings may provide an alternative target for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xiao Wang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Chao Liang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Xin Yao
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Ruo-Han Yang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Zhan-Sheng Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Fan-Ye Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Wen-Qi Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Shu-Hua Pei
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Jing Ma
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Song-Qiang Xie
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China.,Institute of Chemical Biology, School of Pharmacy, Henan University, Kaifeng, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| |
Collapse
|
24
|
Niu J, Li W, Liang C, Wang X, Yao X, Yang RH, Zhang ZS, Liu HF, Liu FY, Pei SH, Li WQ, Sun H, Fang D, Xie SQ. EGF promotes
DKK1
transcription in hepatocellular carcinoma by enhancing the phosphorylation and acetylation of histone H3. Sci Signal 2020; 13:13/657/eabb5727. [DOI: 10.1126/scisignal.abb5727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jie Niu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Wei Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Chao Liang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xiao Wang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xin Yao
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Ruo-Han Yang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Zhan-Sheng Zhang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Han-Fang Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Fan-Ye Liu
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Shu-Hua Pei
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Wen-Qi Li
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Hua Sun
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Dong Fang
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Song-Qiang Xie
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| |
Collapse
|
25
|
Sun D, Zhong J, Wei W, Liu L, Liu J, Lin X. Long non-coding RNAs lnc-ANGPTL1-3:3 and lnc-GJA10-12:1 present as regulators of sentinel lymph node metastasis in breast cancer. Oncol Lett 2020; 20:188. [PMID: 32952657 PMCID: PMC7479524 DOI: 10.3892/ol.2020.12050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) participate in various biological processed involved in tumorigenesis, metastasis and proliferation. The aim of the present study was to identify candidate long non-coding RNAs (lncRNAs) involved in sentinel lymph node (SLN) metastasis in breast cancer. Specimens of SLNs were collected from patients with SLN metastasis via punch biopsy. Total RNA was extracted and RNA sequencing (RNA-seq) was conducted. Differential expression profiles of mRNAs and lncRNAs were obtained via bioinformatics analysis, and Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed on differentially expressed mRNAs. The expression levels of lncRNAs were analyzed via reverse transcription-quantitative PCR (RT-qPCR), and the regulation network of the lncRNAs to downstream microRNAs (miRs) and mRNAs was predicted. Based on RNA-seq results, six differentially expressed candidate lncRNAs were identified in patients with and without SLN metastasis: lnc-ANGPTL1-3:3, lnc-GJA10-12:1, lnc-ACAN-2:1, lnc-ZPBP2-4:1, lnc-GATA3-16:1 and lnc-ACOX3-5:1. KEGG and GO analysis identified that the mitogen-activated protein kinase (MAPK) and PI3K/Akt signaling pathways were the most enriched pathways. After RT-qPCR analysis, lnc-ANGPTL1-3:3 and lnc-GJA10-12:1 exhibited expression patterns that were consistent with those from RNA-seq. Moreover, receiver operating characteristic curve analysis demonstrated that lnc-ANGPTL1-3:3 and lnc-GJA10-12:1 expression levels had high sensitivity and specificity in the diagnosis of SLN metastasis, and that their expression levels were upregulated in patients with axillary lymph node metastasis. Further analysis revealed that lnc-GJA10-12:1 and lnc-ANGPTL1-3:3 were commonly involved in regulating the miR-302 family, including miR-302d-3p and miR-302c-3p, which together targeted AKT1. Additionally, lnc-ANGPTL1-3:3 was predicted to target miR-520b to regulate MAP3K2 expression. lnc-GJA10-12:1 was also predicted to target miR-34a-5p to regulate MAP2K1 and MAP3K9 expression levels, as well as miR-449a to regulate MAP2K1 expression. The results of the present study suggested that lnc-ANGPTL1-3:3 and lnc-GJA10-12:1 may potentially serve a role in SLN metastasis of breast cancer by regulating the PI3K/Akt and MAPK signaling pathways via targeting the miR-302 family, miR-520a-3p, miR-34a-5p and miR-449a. Thus, lnc-ANGPTL1-3:3 and lnc-GJA10-12:1 in SLN may serve as potential markers of breast cancer metastasis.
Collapse
Affiliation(s)
- Desheng Sun
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Jieyu Zhong
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Wei Wei
- Department of Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Li Liu
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Jun Liu
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| | - Xiaona Lin
- Department of Ultrasonography, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
26
|
LncRNA EIF3J-AS1 enhanced esophageal cancer invasion via regulating AKT1 expression through sponging miR-373-3p. Sci Rep 2020; 10:13969. [PMID: 32811869 PMCID: PMC7434778 DOI: 10.1038/s41598-020-70886-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/05/2020] [Indexed: 12/27/2022] Open
Abstract
Esophageal cancer (ECa) remains a major cause of mortality across the globe. The expression of EIF3J-AS1 is altered in a plethora of tumors, but its role in ECa development and progression are undefined. Here, we show that EIF3J-AS1 is up-regulated in ECa and that its expression correlates with advanced TNM stage (P = 0.014), invasion depth (P = 0.001), positive lymph node metastasis (P < 0.001) and poor survival (OS: P = 0.0059; DFS: P = 0.0037) in ECa. Functional experiments showed that knockdown EIF3J-AS1 inhibited ECa growth and metastasis through in vitro and in vivo experiments. Regarding the mechanism, EIF3J-AS1/miR-373-3p/AKT1 established the ceRNA network involved in the modulation of cell progression of ECa cells. Overall, EIF3J-AS1 may exhibit an oncogenic function in ECa via acting as a sponge for miR-373-3p to up-regulate AKT1 mRNA level, and may serve as a potential therapeutic target and a prognostic biomarker for ECa patients.
Collapse
|
27
|
Liao F, Chen L, Luo P, Jiang Z, Chen Z, Wang Z, Zhang C, Wang Y, He J, Wang Q, Wang Y, Liu L, Huang Y, Wang H, Jiang Q, Luo M, Gan Y, Liu Y, Wang Y, Wu J, Xie W, Cheng Z, Dai Y, Li J, Liu Z, Yang F, Shi C. PC4 serves as a negative regulator of skin wound healing in mice. BURNS & TRAUMA 2020; 8:tkaa010. [PMID: 32373645 PMCID: PMC7198317 DOI: 10.1093/burnst/tkaa010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/29/2020] [Indexed: 01/13/2023]
Abstract
Background Human positive cofactor 4 (PC4) was initially characterized as a multifunctional transcriptional cofactor, but its role in skin wound healing is still unclear. The purpose of this study was to explore the role of PC4 in skin wound healing through PC4 knock-in mouse model. Methods A PC4 knock-in mouse model (PC4+/+) with a dorsal full-thickness wound was used to investigate the biological functions of PC4 in skin wound healing. Quantitative PCR, Western blot analysis and immunohistochemistry were performed to evaluate the expression of PC4; Sirius red staining and immunofluorescence were performed to explore the change of collagen deposition and angiogenesis. Proliferation and apoptosis were detected using Ki67 staining and TUNEL assay. Primary dermal fibroblasts were isolated from mouse skin to perform cell scratch experiments, cck-8 assay and colony formation assay. Results The PC4+/+ mice were fertile and did not display overt abnormalities but showed an obvious delay in cutaneous healing of dorsal skin. Histological staining showed insufficient re-epithelialization, decreased angiogenesis and collagen deposition, increased apoptosis and decreased cell proliferation in PC4+/+ skin. Our data also showed decreased migration rate and proliferation ability in cultured primary fibroblasts from PC4+/+ mice in vitro. Conclusions This study suggests that PC4 might serve as a negative regulator of skin wound healing in mice.
Collapse
Affiliation(s)
- Fengying Liao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Long Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhongyong Jiang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chi Zhang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jintao He
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Clinical Medicine, Southwest Medical University, 646000 Luzhou, China
| | - Yawei Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lang Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025 Guiyang, China
| | - Yu Huang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025 Guiyang, China
| | - Huilan Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Clinical Medicine, Southwest Medical University, 646000 Luzhou, China
| | - Qingzhi Jiang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Institute of Clinical Medicine, Southwest Medical University, 646000 Luzhou, China
| | - Min Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.,Department of Toxicology, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, 550025 Guiyang, China
| | - Yibo Gan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yunsheng Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yang Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Wu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wentao Xie
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhuo Cheng
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yali Dai
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jialun Li
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zujuan Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fan Yang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
28
|
Antihypertensive Activity of Eucommia Ulmoides Oliv: Male Flower Extract in Spontaneously Hypertensive Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6432173. [PMID: 32419815 PMCID: PMC7210558 DOI: 10.1155/2020/6432173] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
Eucommia ulmoides Oliv. is a traditional medical plant in Asia; however, it is still unknown whether Eucommia male flowers have an antihypertensive activity. In this study, we found that the aqueous extract of Eucommia ulmoides Oliv. male flowers can lower the blood pressure of SHR in a dose-dependent manner. Mechanistic studies suggested that the aqueous extract of male flowers can promote the mRNA and protein expressions of ACE2 in the kidney of SHR. ELISA assay showed that the plasma levels of ANG II was decreased, while ANG-(1–7) was increased in SHR treated with the aqueous extract of male flowers. ACE2 inhibitor DX600 can reverse the aqueous extract of Eucommia ulmoides Oliv. male flower-induced downregulation of Ang II and upregulation of Ang-(1–7), as well as the reduction of blood pressure in SHR. Moreover, Ang-(1–7)-Mas receptor antagonist A-779 abolished the antihypertensive effects of the aqueous extract of Eucommia ulmoides Oliv. male flower in SHR. The aqueous extract of Eucommia ulmoides Oliv. male flowers exhibited an antihypertensive action through the activation of ACE2-Ang-(1–7)-Mas signaling pathways in spontaneously hypertensive rats.
Collapse
|
29
|
Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Commun Signal 2019; 17:154. [PMID: 31752925 PMCID: PMC6873690 DOI: 10.1186/s12964-019-0450-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AKT, also known as protein kinase B, is a key element of the PI3K/AKT signaling pathway. Moreover, AKT regulates the hallmarks of cancer, e.g. tumor growth, survival and invasiveness of tumor cells. After AKT was discovered in the early 1990s, further studies revealed that there are three different AKT isoforms, namely AKT1, AKT2 and AKT3. Despite their high similarity of 80%, the distinct AKT isoforms exert non-redundant, partly even opposing effects under physiological and pathological conditions. Breast cancer as the most common cancer entity in women, frequently shows alterations of the PI3K/AKT signaling. MAIN CONTENT A plethora of studies addressed the impact of AKT isoforms on tumor growth, metastasis and angiogenesis of breast cancer as well as on therapy response and overall survival in patients. Therefore, this review aimed to give a comprehensive overview about the isoform-specific effects of AKT in breast cancer and to summarize known downstream and upstream mechanisms. Taking account of conflicting findings among the studies, the majority of the studies reported a tumor initiating role of AKT1, whereas AKT2 is mainly responsible for tumor progression and metastasis. In detail, AKT1 increases cell proliferation through cell cycle proteins like p21, p27 and cyclin D1 and impairs apoptosis e.g. via p53. On the downside AKT1 decreases migration of breast cancer cells, for instance by regulating TSC2, palladin and EMT-proteins. However, AKT2 promotes migration and invasion most notably through regulation of β-integrins, EMT-proteins and F-actin. Whilst AKT3 is associated with a negative ER-status, findings about the role of AKT3 in regulation of the key properties of breast cancer are sparse. Accordingly, AKT1 is mutated and AKT2 is amplified in some cases of breast cancer and AKT isoforms are associated with overall survival and therapy response in an isoform-specific manner. CONCLUSIONS Although there are several discussed hypotheses how isoform specificity is achieved, the mechanisms behind the isoform-specific effects remain mostly unrevealed. As a consequence, further effort is necessary to achieve deeper insights into an isoform-specific AKT signaling in breast cancer and the mechanism behind it.
Collapse
Affiliation(s)
- Nico Hinz
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
30
|
Effect of 6-Shogaol on the Glucose Uptake and Survival of HT1080 Fibrosarcoma Cells. Pharmaceuticals (Basel) 2019; 12:ph12030131. [PMID: 31505728 PMCID: PMC6789756 DOI: 10.3390/ph12030131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/10/2019] [Accepted: 09/03/2019] [Indexed: 12/29/2022] Open
Abstract
Ginger is a plant that is native to southern China. In the last decade and research on the components of ginger has significantly increased; of these components, 6-shogaol exhibits the greatest potential antitumor capacity. However, the molecular mechanism through which 6-shogaol exerts its effects has not yet been elucidated. In this study, the effect of 6-shogaol on tumor cells that were derived from human fibrosarcoma (HT1080) was evaluated. Cell viability was determined by a (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) MTT assay testing different concentrations of 6-shogaol (2.5–150 μM). Subsequently, the effect of 6-shogaol on reactive oxygen species (ROS) production, glucose uptake, and protein expression of the signaling pathway phosphatase and tensin homolog/ protein kinase B /mammalian target of rapamycin (PTEN/Akt/mTOR) was measured. 6-Shogaol reduced the viability of the tumor cells and caused an increase in ROS production, which was attenuated with the addition of N-acetylcysteine, and the recovery of cell viability was observed. The increase in ROS production in response to 6-shogaol was associated with cell death. Similarly, glucose uptake decreased with incremental concentrations of 6-shogaol, and an increase in the expression of mTOR-p and Akt-p proteins was observed; PTEN was active in all the treatments with 6-shogaol. Thus, the results suggest that cells activate uncontrolled signaling pathways, such as phosphoinositide 3-kinase (PI3K)/Akt/mTOR, among other alternative mechanisms of metabolic modulation and of survival in order to counteract the pro-oxidant effect of 6-shogaol and the decrease in glucose uptake. Interestingly, a differential response was observed when non-cancerous cells were treated with 6-shogaol.
Collapse
|
31
|
Cao J, Zhang Y, Wu L, Shan L, Sun Y, Jiang X, Tao J. Electrical stimulation of the superior sagittal sinus suppresses A-type K + currents and increases P/Q- and T-type Ca 2+ currents in rat trigeminal ganglion neurons. J Headache Pain 2019; 20:87. [PMID: 31375062 PMCID: PMC6734278 DOI: 10.1186/s10194-019-1037-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/28/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Migraine is a debilitating neurological disorder involving abnormal trigeminovascular activation and sensitization. However, the underlying cellular and molecular mechanisms remain unclear. METHODS A rat model of conscious migraine was established through the electrical stimulation (ES) of the dural mater surrounding the superior sagittal sinus. Using patch clamp recording, immunofluorescent labelling, enzyme-linked immunosorbent assays and western blot analysis, we studied the effects of ES on sensory neuronal excitability and elucidated the underlying mechanisms mediated by voltage-gated ion channels. RESULTS The calcitonin gene-related peptide (CGRP) level in the jugular vein blood and the number of CGRP-positive neurons in the trigeminal ganglia (TGs) were significantly increased in rats with ES-induced migraine. The application of ES increased actional potential firing in both small-sized IB4-negative (IB4-) and IB4+ TG neurons. No significant changes in voltage-gated Na+ currents were observed in the ES-treated groups. ES robustly suppressed the transient outward K+ current (IA) in both types of TG neurons, while the delayed rectifier K+ current remained unchanged. Immunoblot analysis revealed that the protein expression of Kv4.3 was significantly decreased in the ES-treated groups, while Kv1.4 remained unaffected. Interestingly, ES increased the P/Q-type and T-type Ca2+ currents in small-sized IB4- TG neurons, while there were no significant changes in the IB4+ subpopulation of neurons. CONCLUSION These results suggest that ES decreases the IA in small-sized TG neurons and increases P/Q- and T-type Ca2+ currents in the IB4- subpopulation of TG neurons, which might contribute to neuronal hyperexcitability in a rat model of ES-induced migraine.
Collapse
Affiliation(s)
- Junping Cao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China.,Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yuan Zhang
- Department of Geriatrics & Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, People's Republic of China
| | - Lei Wu
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Lidong Shan
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Yufang Sun
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Xinghong Jiang
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China
| | - Jin Tao
- Department of Physiology and Neurobiology & Centre for Ion Channelopathy, Medical College of Soochow University, 199 Ren-Ai Road, Suzhou, 215123, People's Republic of China. .,Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, People's Republic of China.
| |
Collapse
|
32
|
Wu Z, Wu J, Zhao Q, Fu S, Jin J. Emerging roles of aerobic glycolysis in breast cancer. Clin Transl Oncol 2019; 22:631-646. [PMID: 31359335 DOI: 10.1007/s12094-019-02187-8] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022]
Abstract
Altered aerobic glycolysis is a well-recognized characteristic of cancer cell energy metabolism, known as the Warburg effect. Even in the presence of abundant oxygen, a majority of tumor cells produce substantial amounts of energy through a high glycolytic metabolism, and breast cancer (BC) is no exception. Breast cancer continues to be the second leading cause of cancer-associated mortality in women worldwide. However, the precise role of aerobic glycolysis in the development of BC remains elusive. Therefore, the present review attempts to address the implication of key enzymes of the aerobic glycolytic pathway including hexokinase (HK), phosphofructokinase (PFK) and pyruvate kinase (PK), glucose transporters (GLUTs), together with related signaling pathways including protein kinase B(PI3K/AKT), mammalian target of rapamycin (mTOR) and adenosine monophosphate-activated protein kinase (AMPK) and transcription factors (c-myc, p53 and HIF-1) in the research of BC. Thus, the review of aerobic glycolysis in BC may evoke novel ideas for the BC treatment.
Collapse
Affiliation(s)
- Z Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - J Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Q Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, People's Republic of China
| | - S Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - J Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
33
|
Niu J, Li XM, Wang X, Liang C, Zhang YD, Li HY, Liu FY, Sun H, Xie SQ, Fang D. DKK1 inhibits breast cancer cell migration and invasion through suppression of β-catenin/MMP7 signaling pathway. Cancer Cell Int 2019; 19:168. [PMID: 31285694 PMCID: PMC6591985 DOI: 10.1186/s12935-019-0883-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Background DKK1 has been reported to act as a tumor suppressor in breast cancer. However, the mechanism of DKK1 inhibits breast cancer migration and invasion was still unclear. Methods Western blot and real time PCR was used to detect the expression of DKK1, β-catenin and MMP7 in breast cancer cells. Wound scratch assay and transwell assay was employed to examine migration and invasion of breast cancer cell. Results DKK1 overexpression dramatically inhibits breast cancer cell migration and invasion. Knockdown of DKK1 promotes migration and invasion of breast cancer cells. DKK1 suppressed breast cancer cell migration and invasion through suppression of β-catenin and MMP7 expression. XAV-939, an inhibitor of β-catenin accumulation could reverse DKK1 silencing-induced MMP7 expression in breast cancer cells. Meanwhile, XAV-939 also could reverse the increase in the cell number invaded through Matrigel when DKK1 was knockdown. Furthermore, depletion of MMP7 also could reverse DKK1 knockdown-induced increase in the cell number invaded through Matrigel. Conclusions DKK1 inhibits migration and invasion of breast cancer cell through suppression of β-catenin/MMP7 pathway, our findings offered a potential alternative for breast cancer prevention and treatment.
Collapse
Affiliation(s)
- Jie Niu
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Xiao-Meng Li
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Xiao Wang
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Chao Liang
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Yi-Dan Zhang
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Hai-Ying Li
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Fan-Ye Liu
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Hua Sun
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Song-Qiang Xie
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China.,2Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| | - Dong Fang
- 1Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004 China
| |
Collapse
|
34
|
Xu C, Liu F, Xiang G, Cao L, Wang S, Liu J, Meng Q, Xu D, Lv S, Jiao J, Niu Y. β-Catenin nuclear localization positively feeds back on EGF/EGFR-attenuated AJAP1 expression in breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:238. [PMID: 31171012 PMCID: PMC6554977 DOI: 10.1186/s13046-019-1252-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/28/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Adherent junction associated protein 1 (AJAP1), a typical molecule of adherent junctions, has been found to be a tumor suppressor in many cancer types. Aberrant activation of β-catenin has been demonstrated to be associated with malignant biological properties of tumors including breast cancer. This study aimed to investigate the function and mechanism of AJAP1-mediated β-catenin activity of breast cancer lines in vitro and in breast cancer patients. METHODS AJAP1 and β-catenin expressions in breast cancer tissues and cell lines were detected by immunohistochemistry, western blotting and qRT-PCR. The EGF/EGFR axis-mediated AJAP1 attenuated β-catenin nuclear location was measured by western blotting, immunofluorescence assay, co-immunoprecipitation, luciferase assay and ubiquitination assays. Furthermore, the function of AJAP1 and β-catenin regulated breast cancer progression was explored both in vivo and in vitro. RESULTS It was found that AJAP1 had a high negative correlation with β-catenin nuclear expression and was a novel tumor suppressor in breast cancer. AJAP1 loss can mediate β-catenin accumulated in cytoplasm and then transferred it to the nucleus, activating β-catenin transcriptional activity and downstream genes. Additionally, β-catenin can reverse the invasion, proliferation ability and tumorigenicity of the depletion of AJAP1 caused both in vivo and in vitro. Besides, EGF/EGFR also involved in the process of AJAP1-depiction induced β-catenin transactivation to the nucleus. More importantly, EGFR depletion/AJAP1 knocked down promoted the progression of breast cancer by regulating the activity of β-catenin nuclear transactivation. CONCLUSION This study demonstrated that AJAP1 acted as a putative tumor suppressor while β-catenin nuclear localization positively fed back on EGF/EGFR-attenuated AJAP1 expression in breast cancer, which might be beneficial to develop new therapeutic targets for decreasing nuclear β-catenin-mediated malignancy in breast cancer.
Collapse
Affiliation(s)
- Cong Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fang Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guomin Xiang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lu Cao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shuling Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jing Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qingxiang Meng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Danni Xu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shuhua Lv
- Department of Pathology, Tianjin Union Medical Center, Tianjin People's Hospital, Tianjin, 300121, China
| | - Jiao Jiao
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China.,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yun Niu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China. .,Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, West Huanhu Road, Ti Yuan Bei, Hexi District, Tianjin, 300060, Tianjin, China. .,Department of Breast Cancer Pathology and Research Laboratory, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
35
|
Niu J, Wang X, Liang C, Zhang YD, Liu FY, Li HY, Xie SQ, Sun H, Fang D. Suppression of epidermal growth factor receptor-mediated β-catenin nuclear accumulation enhances the anti-tumor activity of phosphoinositide 3-kinase inhibitor in breast cancer. Cell Biol Int 2019; 43:931-939. [PMID: 31124219 DOI: 10.1002/cbin.11183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/04/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) signaling is frequently deregulated in breast cancer and plays a critical role in tumor progression. However, resistance to PI3K inhibitors in breast cancer has emerged, which is due to the enhanced β-catenin nuclear accumulation. Until now, the mechanisms underlying PI3K inhibition-induced β-catenin nuclear accumulation remains largely unknown. In the present study, we found inhibition of PI3K with LY294002 promoted β-catenin nuclear accumulation in MCF-7 and MDA-MB-231 breast cancer cells. Combining PI3K inhibitor LY294002 with XAV-939, an inhibitor against β-catenin nuclear accumulation, produced an additive anti-proliferation effect against breast cancer cells. Subsequent experiments suggested β-catenin nuclear accumulation induced by PI3K inhibition depended on the feedback activation of epidermal growth factor receptor (EGFR) signaling pathway in breast cancer cells. Inhibition of EGFR phosphorylation with Gefitinib enhanced anti-proliferation effect of PI3K inhibitor LY294002 in MCF-7 and MDA-MB-231 cells. Taken together, our findings may elucidate a possible mechanism explaining the poor outcome of PI3K inhibitors in breast cancer treatment.
Collapse
Affiliation(s)
- Jie Niu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Xiao Wang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Chao Liang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Yi-Dan Zhang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Fan-Ye Liu
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Hai-Ying Li
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Song-Qiang Xie
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China.,School of Pharmacy, Institute of Chemical Biology, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Hua Sun
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| | - Dong Fang
- School of Pharmacy, Institute for Innovative Drug Design and Evaluation, Henan University, N. Jinming Ave, 475004 Kaifeng, China
| |
Collapse
|
36
|
Luo P, Zhang C, Liao F, Chen L, Liu Z, Long L, Jiang Z, Wang Y, Wang Z, Liu Z, Miao H, Shi C. Transcriptional positive cofactor 4 promotes breast cancer proliferation and metastasis through c-Myc mediated Warburg effect. Cell Commun Signal 2019; 17:36. [PMID: 30992017 PMCID: PMC6469038 DOI: 10.1186/s12964-019-0348-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/03/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The human positive cofactor 4 (PC4) is initially identified as a transcriptional cofactor and has an important role in embryonic development and malignant transformation. However, the clinical significance and the molecular mechanisms of PC4 in breast cancer development and progression are still unknown. METHODS We investigated PC4 expression in 114 cases of primary breast cancer and matched normal breast tissue specimens, and studied the impact of PC4 expression as well as the molecular mechanisms of this altered expression on breast cancer growth and metastasis both in vitro and in vivo. RESULTS PC4 was significantly upregulated in breast cancer and high PC4 expression was positively correlated with metastasis and poor prognosis of patients. Gene set enrichment analysis (GSEA) demonstrated that the gene sets of cell proliferation and Epithelial-Mesenchymal Transition (EMT) were positively correlated with elevated PC4 expression. Consistently, loss of PC4 markedly inhibited the growth and metastasis of breast cancer both in vitro and in vivo. Mechanistically, PC4 exerted its oncogenic functions by directly binding to c-Myc promoters and inducing Warburg effect. CONCLUSIONS Our study reveals for the first time that PC4 promotes breast cancer progression by directly regulating c-Myc transcription to promote Warburg effect, implying a novel therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Peng Luo
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Chi Zhang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Fengying Liao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Long Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Zhenyu Liu
- Institute of Immunology, Third Military Medical University, Chongqing, 400038 China
| | - Lei Long
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Zhongyong Jiang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Yawei Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Zujuan Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University, Chongqing, 400038 China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, 400038 China
| |
Collapse
|