1
|
Ramakrishnan G, Terry AR, Nogueira V, Magdy A, Hay N. Deletion of AMP-activated protein kinase impairs metastasis and is rescued by ROS scavenging or ectopic CD36 expression. Cell Rep 2025; 44:115183. [PMID: 39798092 DOI: 10.1016/j.celrep.2024.115183] [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: 06/18/2024] [Revised: 11/18/2024] [Accepted: 12/18/2024] [Indexed: 01/15/2025] Open
Abstract
AMPK's role in tumor initiation and progression is controversial. Here, we provide genetic evidence that AMPK is required for metastasis in mouse models of breast cancer. In a mouse model of spontaneous breast cancer metastasis, the deletion of AMPK before and after tumor onset decreased breast cancer metastasis, and similar results were obtained after AMPK deletion in breast cancer cell lines. The deletion of AMPK induces reactive oxygen species (ROS) levels in vitro and lipid oxidation in vivo, which likely impede metastasis. Indeed, antioxidants restore the ability of AMPK-deficient tumors to metastasize. By inhibiting acetyl-coenzyme A (CoA) carboxylases 1 and 2, AMPK maintains NADPH levels by reducing NADPH consumption in fatty acid synthesis and increasing NADPH generation via fatty acid oxidation, thus increasing the dependency on auxotrophic fatty acids. Consistently, AMPK is required for the expression of the fatty acid transporter CD36 in tumors, and ectopic expression of CD36 in AMPK-deficient cells restored their ability to metastasize.
Collapse
Affiliation(s)
- Gopalakrishnan Ramakrishnan
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Alexander R Terry
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Veronique Nogueira
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ahmed Magdy
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA; Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
2
|
Vermehren-Schmaedick A, Peto M, Wagoner W, Chiotti KE, Ramsey E, Wang X, Rakshe S, Minnier J, Sears R, Spellman P, Luoh SW. Mutation of SIVA, a candidate metastasis gene identified from clonally related bilateral breast cancers, promotes breast cancer cell spread in vitro and in vivo. PLoS One 2024; 19:e0302856. [PMID: 38722955 PMCID: PMC11081324 DOI: 10.1371/journal.pone.0302856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/12/2024] [Indexed: 05/13/2024] Open
Abstract
Metastasis is the most dreaded outcome after a breast cancer diagnosis, and little is known regarding what triggers or promotes breast cancer to spread distally, or how to prevent or eradicate metastasis effectively. Bilateral breast cancers are an uncommon form of breast cancers. In our study, a percentage of bilateral breast cancers were clonally related based on copy number variation profiling. Whole exome sequencing and comparative sequence analysis revealed that a limited number of somatic mutations were acquired in this "breast-to-breast" metastasis that might promote breast cancer distant spread. One somatic mutation acquired was SIVA-D160N that displayed pro-metastatic phenotypes in vivo and in vitro. Over-expression of SIVA-D160N promoted migration and invasion of human MB-MDA-231 breast cancer cells in vitro, consistent with a dominant negative interfering function. When introduced via tail vein injection, 231 cells over-expressing SIVA-D160N displayed enhanced distant spread on IVIS imaging. Over-expression of SIVA-D160N promoted invasion and anchorage independent growth of mouse 4T1 breast cancer cells in vitro. When introduced orthotopically via mammary fat pad injection in syngeneic Balb/c mice, over-expression of SIVA-D160N in 4T1 cells increased orthotopically implanted mammary gland tumor growth as well as liver metastasis. Clonally related bilateral breast cancers represented a novel system to investigate metastasis and revealed a role of SIVA-D160N in breast cancer metastasis. Further characterization and understanding of SIVA function, and that of its interacting proteins, may elucidate mechanisms of breast cancer metastasis, providing clinically useful biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Veterans Administration Portland Health Care System, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Myron Peto
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Wendy Wagoner
- Veterans Administration Portland Health Care System, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Kami E. Chiotti
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Elizabeth Ramsey
- Veterans Administration Portland Health Care System, Portland, Oregon, United States of America
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Xiaoyan Wang
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Shauna Rakshe
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jessica Minnier
- Knight Cancer Institute, Biostatistics Shared Resource, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Rosalie Sears
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
- Brenden-Colson Center for Pancreatic Care, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Paul Spellman
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- University of California Los Angeles, Los Angeles, California, United States of America
| | - Shiuh-Wen Luoh
- Veterans Administration Portland Health Care System, Portland, Oregon, United States of America
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
3
|
Terry AR, Nogueira V, Rho H, Ramakrishnan G, Li J, Kang S, Pathmasiri KC, Bhat SA, Jiang L, Kuchay S, Cologna SM, Hay N. CD36 maintains lipid homeostasis via selective uptake of monounsaturated fatty acids during matrix detachment and tumor progression. Cell Metab 2023; 35:2060-2076.e9. [PMID: 37852255 PMCID: PMC11748917 DOI: 10.1016/j.cmet.2023.09.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 04/11/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
A high-fat diet (HFD) promotes metastasis through increased uptake of saturated fatty acids (SFAs). The fatty acid transporter CD36 has been implicated in this process, but a detailed understanding of CD36 function is lacking. During matrix detachment, endoplasmic reticulum (ER) stress reduces SCD1 protein, resulting in increased lipid saturation. Subsequently, CD36 is induced in a p38- and AMPK-dependent manner to promote preferential uptake of monounsaturated fatty acids (MUFAs), thereby maintaining a balance between SFAs and MUFAs. In attached cells, CD36 palmitoylation is required for MUFA uptake and protection from palmitate-induced lipotoxicity. In breast cancer mouse models, CD36-deficiency induced ER stress while diminishing the pro-metastatic effect of HFD, and only a palmitoylation-proficient CD36 rescued this effect. Finally, AMPK-deficient tumors have reduced CD36 expression and are metastatically impaired, but ectopic CD36 expression restores their metastatic potential. Our results suggest that, rather than facilitating HFD-driven tumorigenesis, CD36 plays a supportive role by preventing SFA-induced lipotoxicity.
Collapse
Affiliation(s)
- Alexander R Terry
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Veronique Nogueira
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Hyunsoo Rho
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Gopalakrishnan Ramakrishnan
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jing Li
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Soeun Kang
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Koralege C Pathmasiri
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sameer Ahmed Bhat
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Liping Jiang
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Shafi Kuchay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Stephanie M Cologna
- Department of Chemistry, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA; Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
4
|
Liu H, Xie T, Liu Y. Ginsenoside Rg3 inhibits the malignant progression of cervical cancer cell by regulating AKT2 expression. Heliyon 2023; 9:e19045. [PMID: 37664735 PMCID: PMC10469050 DOI: 10.1016/j.heliyon.2023.e19045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023] Open
Abstract
Although ginsenoside Rg3 has been shown to exert anticancer effects in various malignancies, the effects and molecular mechanisms of ginsenoside Rg3 in cervical cancer (CC) remain unclear. This study explored the effect of ginsenoside Rg3 on CC development at the cellular level. The effect of ginsenoside Rg3 on cell proliferation was measured using colony formation and Cell Counting Kit-8 assays. Migration, invasion, and in vitro angiogenesis of CC cells were detected using wound healing, transwell, and tube formation assays, respectively. In addition, we explored the target genes and molecular mechanisms of ginsenoside Rg3 in CC cells overexpressing AKT serine/threonine kinase 2 (AKT2). The results indicated that ginsenoside Rg3 suppressed proliferation, migration, invasion, and tube formation of CC cells in vitro. In addition, ginsenoside Rg3 treatment decreased the expression of AKT2 in CC cells. Moreover, ginsenoside Rg3 treatment partially reversed AKT2 overexpression-mediated reduction in cell proliferation, migration, invasion, and tube formation. In conclusion, the above findings suggested that ginsenoside Rg3 inhibits CC progression via regulation of AKT2 expression, which might provide a potential therapeutic target for tumor therapy.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pharmacy, Shangqiu First People's Hospital, Shangqiu, 476000, China
| | - Tingting Xie
- Department of Pharmacy, Medical Supplies Center of Chinese PLA General hospital, Beijing, 10000, China
| | - Yuan Liu
- Department of Pharmacy, Shangqiu First People's Hospital, Shangqiu, 476000, China
| |
Collapse
|
5
|
Wu L, Xiao H, Hong Y, Xie M, Yu Y, Jiang L. CircRNA Circ_0000118 Regulates Malignancy of Cervical Cancer Cells by Regulating miR-211-5p/miR-377-3p/AKT2 Axis. Biochem Genet 2023; 61:1625-1644. [PMID: 36719624 PMCID: PMC10371915 DOI: 10.1007/s10528-023-10332-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 01/06/2023] [Indexed: 02/01/2023]
Abstract
CircRNAs are implicated in the development of several cancers. Nevertheless, the involvement of circ_0000118 in the development of cervical cancer (CC) remains unclear. Circ_0000118 levels in tumor tissues and cells were examined by qRT-PCR. The function of circ_0000118 in regulating the malignancy of CC cells was investigated using functional assays, including CCK-8, colony formation, transwell, and tube formation experiments. The functional interaction between circ_0000118 and microRNAs were validated by dual-luciferase activity assay and RNA precipitation experiments. In vivo mouse model was employed to assess the effect of circ_0000118 in the tumorigenesis of CC cells. Circ_0000118 was overexpressed in CC cells and tissues. Loss-of-function experiments demonstrated that circ_0000118 knockdown impaired the proliferation and tumor sphere formation, as well as the angiogenic potential of CC cells. RNA interaction experiments confirmed that circ_0000118 sponged miR-211-5p and miR-377-3p. AKT2 was found to be a target gene negatively modulated by miR-211-5p and miR-377-3p. AKT2 overexpression rescued the inhibition of circ_0000118 downregulation on CC cells. Our study suggested that circ_0000118 functions as an oncogenic factor in progression of CC by maintaining AKT2 level through targeting miR-211-5p and miR-377-3p as a ceRNA (competitive endogenous RNA), which provides novel therapeutic target in the management of CC.
Collapse
Affiliation(s)
- Lilan Wu
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China
| | - Huiqin Xiao
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China
| | - Yaqin Hong
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China
| | - Meihua Xie
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China
| | - Yanxia Yu
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China
| | - Lijuan Jiang
- Department of Obstetrics and Gynecology, Chunan County Traditional Chinese Medicine Hospital, No.1 Xin'an West Road, Chun'an, Hangzhou, 311700, Zhejiang, China.
| |
Collapse
|
6
|
Li P, Hao X, Liu J, Zhang Q, Liang Z, Li X, Liu H. miR-29a-3p Regulates Autophagy by Targeting Akt3-Mediated mTOR in SiO 2-Induced Lung Fibrosis. Int J Mol Sci 2023; 24:11440. [PMID: 37511199 PMCID: PMC10380316 DOI: 10.3390/ijms241411440] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/02/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Silicosis is a refractory pneumoconiosis of unknown etiology that is characterized by diffuse lung fibrosis, and microRNA (miRNA) dysregulation is connected to silicosis. Emerging evidence suggests that miRNAs modulate pulmonary fibrosis through autophagy; however, its underlying molecular mechanism remains unclear. In agreement with miRNA microarray analysis, the qRT-PCR results showed that miR-29a-3p was significantly decreased in the pulmonary fibrosis model both in vitro and in vivo. Increased autophagosome was observed via transmission electron microscopy in lung epithelial cell models and lung tissue of silicosis mice. The expression of autophagy-related proteins LC3α/β and Beclin1 were upregulated. The results from using 3-methyladenine, an autophagy inhibitor, or rapamycin, an autophagy inducer, together with TGF-β1, indicated that autophagy attenuates fibrosis by protecting lung epithelial cells. In TGF-β1-treated TC-1 cells, transfection with miR-29a-3p mimics activated protective autophagy and reduced alpha-smooth muscle actin and collagen I expression. miRNA TargetScan predicted, and dual-luciferase reporter experiments identified Akt3 as a direct target of miR-29a-3p. Furthermore, Akt3 expression was significantly elevated in the silicosis mouse model and TGF-β1-treated TC-1 cells. The mammalian target of rapamycin (mTOR) is a central regulator of the autophagy process. Silencing Akt3 inhibited the transduction of the mTOR signaling pathway and activated autophagy in TGF-β1-treated TC-1 cells. These results show that miR-29a-3p overexpression can partially reverse the fibrotic effects by activating autophagy of the pulmonary epithelial cells regulated by the Akt3/mTOR pathway. Therefore, targeting miR-29a-3p may provide a new therapeutic strategy for silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Peiyuan Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
| | - Jiaxin Liu
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Qinxin Zhang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Zixuan Liang
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Xinran Li
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (P.L.); (J.L.); (Q.Z.); (Z.L.); (X.L.)
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan 063210, China
| |
Collapse
|
7
|
Anifowose LO, Paimo OK, Adegboyega FN, Ogunyemi OM, Akano RO, Hammad SF, Ghazy MA. Molecular docking appraisal of Dysphania ambrosioides phytochemicals as potential inhibitor of a key triple-negative breast cancer driver gene. In Silico Pharmacol 2023; 11:15. [PMID: 37323538 PMCID: PMC10267046 DOI: 10.1007/s40203-023-00152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a lethal and aggressive breast cancer subtype. It is characterized by the deficient expression of the three main receptors implicated in breast cancers, making it unresponsive to hormone therapy. Hence, an existing need to develop a targeted molecular therapy for TNBC. The PI3K/AKT/mTOR signaling pathway mediates critical cellular processes, including cell proliferation, survival, and angiogenesis. It is activated in approximately 10-21% of TNBCs, emphasizing the importance of this intracellular target in TNBC treatment. AKT is a prominent driver of the PI3K/AKT/mTOR pathway, validating it as a promising therapeutic target. Dysphania ambrosioides is an important ingredient of Nigeria's traditional herbal recipe for cancer treatment. Thus, our present study explores its anticancer properties through a structure-based virtual screening of 25 biologically active compounds domiciled in the plant. Interestingly, our molecular docking study identified several potent inhibitors of AKT 1 and 2 isoforms from D. ambrosioides. However, cynaroside and epicatechin gallate having a binding energy of - 9.9 and - 10.2 kcal/mol for AKT 1 and 2, respectively, demonstrate considerable drug-likeness than the reference drug (capivasertib), whose respective binding strengths for AKT 1 and 2 are - 9.5 and - 8.4 kcal/mol. Lastly, the molecular dynamics simulation experiment showed that the simulated complex systems of the best hits exhibit structural stability throughout the 50 ns run. Together, our computational modeling analysis suggests that these compounds could emerge as efficacious drug candidates in the treatment of TNBC. Nevertheless, further experimental, translational, and clinical research is required to establish an empirical clinical application. Graphical Abstract A structure-based virtual screening and simulation of Dysphania ambrosioides phytochemicals in the active pocket of AKT 1 and 2 isoforms.
Collapse
Affiliation(s)
- Lateef O. Anifowose
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State Nigeria
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State Nigeria
- Department of Biotechnology, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| | - Oluwatomiwa K. Paimo
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State Nigeria
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Ogun State Nigeria
| | - Fikayo N. Adegboyega
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State Nigeria
- Department of Biotechnology, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| | - Oludare M. Ogunyemi
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State Nigeria
| | - Rukayat O. Akano
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State Nigeria
| | - Sherif F. Hammad
- Department of Biotechnology, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| | - Mohamed A. Ghazy
- Department of Biotechnology, Institute of Basic and Applied Sciences, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| |
Collapse
|
8
|
Huang H, Ke C, Zhang D, Wu J, Zhang P. Molecular mechanism study and tumor heterogeneity of Chinese angelica and Fructus aurantii in the treatment of colorectal cancer through computational and molecular dynamics. Funct Integr Genomics 2023; 23:106. [PMID: 36977932 DOI: 10.1007/s10142-023-01042-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE Screening Chinese angelica (CHA) and Fructus aurantii (FRA) for ingredients with therapeutic effects on colorectal cancer (CRC) and discovering novel targets for the prevention or treatment of CRC. METHODS TCMSP database as a starting point for the initial selection of ingredients and targets, we screened and validated the ingredients and targets of CHA and FRA using tools such as Autodock Vina, R 4.2.0, and GROMACS. To obtain the pharmacokinetic information of the active ingredients, we performed ADMET prediction and consulted a large number of works related to CRC cell lines for the discussion and validation of the results. RESULTS Molecular dynamics simulation results showed the complexes formed between these components and targets can exist in a very stable tertiary structure under the human environment, and their side effects can be ignored. CONCLUSIONS Our study successfully explains the effective mechanism of CHA and FRA for improving CRC while predicting the potential targets PPARG, AKT1, RXRA, and PPARA of CHA and FRA for CRC treatment, which provides a new foundation for investigating the novel compounds of TCMs and a new direction for subsequent CRC research.
Collapse
Affiliation(s)
- He Huang
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chunlian Ke
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Dongdong Zhang
- School of Life Sciences, Shihezi University, Shihezi, 832003, China.
| | - Jiezhong Wu
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Peng Zhang
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| |
Collapse
|
9
|
Huang Z, Deng T. miR-204-3p Regulates Glioma Cell Biological Behaviors via Targeting Protein Kinase B (AKT1). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study assesses miR-204-3p’s role in glioma. Cells were transfected with miR-204-3pmimics, miR-204-3p inhibitor, or si-AKT1 to measure cell proliferation, invasion, migration and apoptosis. Glioma tissues showed a significantly downregulated miR-204-3p, whose knockdown can
significantly promote cell proliferation, migration and invasion. However, all the above changes or cell behavior were inhibited by overexpression of miR-204-3p. miR-204-3p regulated AKT1 and its silence can promote cell proliferation and decrease apoptosis by increasing AKT1 expression. However,
si-AKT1 transfection inhibited cell proliferation and promote apoptosis induced by miR-204-3p knockdown. In summary, miR-204-3p regulates glioma cell biological behaviors by targeting AKT1.
Collapse
Affiliation(s)
- Zhengbiao Huang
- Department of Neurology, Hubei Yichang Guoyao Gezhouba Central Hospital, Yichang, 443002, Hubei, China
| | - Tianling Deng
- Department of Neurology, Hubei Yichang Guoyao Gezhouba Central Hospital, Yichang, 443002, Hubei, China
| |
Collapse
|
10
|
Tsai PJ, Lai YH, Manne RK, Tsai YS, Sarbassov D, Lin HK. Akt: a key transducer in cancer. J Biomed Sci 2022; 29:76. [PMID: 36180910 PMCID: PMC9526305 DOI: 10.1186/s12929-022-00860-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/21/2022] [Indexed: 01/27/2023] Open
Abstract
Growth factor signaling plays a pivotal role in diverse biological functions, such as cell growth, apoptosis, senescence, and migration and its deregulation has been linked to various human diseases. Akt kinase is a central player transmitting extracellular clues to various cellular compartments, in turn executing these biological processes. Since the discovery of Akt three decades ago, the tremendous progress towards identifying its upstream regulators and downstream effectors and its roles in cancer has been made, offering novel paradigms and therapeutic strategies for targeting human diseases and cancers with deregulated Akt activation. Unraveling the molecular mechanisms for Akt signaling networks paves the way for developing selective inhibitors targeting Akt and its signaling regulation for the management of human diseases including cancer.
Collapse
Affiliation(s)
- Pei-Jane Tsai
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsin Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Rajesh Kumar Manne
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yau-Sheng Tsai
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Dos Sarbassov
- Biology Department, School of Sciences and Humanities, and National Laboratory Astana, Nazarbayev University, Nur-Sultan City, 010000, Kazakhstan.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
11
|
Kopanja D, Chand V, O’Brien E, Mukhopadhyay NK, Zappia MP, Islam AB, Frolov MV, Merrill BJ, Raychaudhuri P. Transcriptional Repression by FoxM1 Suppresses Tumor Differentiation and Promotes Metastasis of Breast Cancer. Cancer Res 2022; 82:2458-2471. [PMID: 35583996 PMCID: PMC9258028 DOI: 10.1158/0008-5472.can-22-0410] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/07/2023]
Abstract
UNLABELLED The transcription factor Forkhead box M1 (FoxM1) is overexpressed in breast cancers and correlates with poor prognosis. Mechanistically, FoxM1 associates with CBP to activate transcription and with Rb to repress transcription. Although the activating function of FoxM1 in breast cancer has been well documented, the significance of its repressive activity is poorly understood. Using CRISPR-Cas9 engineering, we generated a mouse model that expresses FoxM1-harboring point mutations that block binding to Rb while retaining its ability to bind CBP. Unlike FoxM1-null mice, mice harboring Rb-binding mutant FoxM1 did not exhibit significant developmental defects. The mutant mouse line developed PyMT-driven mammary tumors that were deficient in lung metastasis, which was tumor cell-intrinsic. Single-cell RNA-seq of the tumors revealed a deficiency in prometastatic tumor cells and an expansion of differentiated alveolar type tumor cells, and further investigation identified that loss of the FoxM1/Rb interaction caused enhancement of the mammary alveolar differentiation program. The FoxM1 mutant tumors also showed increased Pten expression, and FoxM1/Rb was found to activate Akt signaling by repressing Pten. In human breast cancers, expression of FoxM1 negatively correlated with Pten mRNA. Furthermore, the lack of tumor-infiltrating cells in FoxM1 mutant tumors appeared related to decreases in pro-metastatic tumor cells that express factors required for infiltration. These observations demonstrate that the FoxM1/Rb-regulated transcriptome is critical for the plasticity of breast cancer cells that drive metastasis, identifying a prometastatic role of Rb when bound to FoxM1. SIGNIFICANCE This work provides new insights into how the interaction between FoxM1 and Rb facilitates the evolution of metastatic breast cancer cells by altering the transcriptome.
Collapse
Affiliation(s)
- Dragana Kopanja
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Vaibhav Chand
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Eilidh O’Brien
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nishit K. Mukhopadhyay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Maria P. Zappia
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Abul B.M.M.K. Islam
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Maxim V. Frolov
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Bradley J. Merrill
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Pradip Raychaudhuri
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
- Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
12
|
Liu H, Li X, Li H, Feng L, Sun G, Sun G, Wu L, Hu Y, Liu L, Wang H. Potential molecular mechanisms and clinical progress in liver metastasis of breast cancer. Biomed Pharmacother 2022; 149:112824. [PMID: 35306430 DOI: 10.1016/j.biopha.2022.112824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women and the leading cause of cancer death in women. About 30% of breast cancer patients have metastasis every year, which greatly increases the mortality rate of breast cancer. The main target organs for metastasis are bone, brain, liver and lung. The breast cancer liver metastasis (BCLM) mechanism is not fully clarified. This is a complex process involving multiple factors, which is not only related to the microenvironment of the primary tumor and liver, but also regulated by a variety of signaling pathways. Clarifying these mechanisms is of great help to guide clinical treatment. With the in-depth study of BCLM, a variety of new treatment schemes such as targeted therapy and endocrine therapy provide new ideas for the cure of BCLM. In this review, we will summarize the molecular mechanism and treatment of BCLM.
Collapse
Affiliation(s)
- Hanyuan Liu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyang Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Feng
- School of Public Health, Fudan University, Shanghai, China
| | - Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Hu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Li Liu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Hanjin Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
13
|
A non-catalytic scaffolding activity of hexokinase 2 contributes to EMT and metastasis. Nat Commun 2022; 13:899. [PMID: 35173161 PMCID: PMC8850586 DOI: 10.1038/s41467-022-28440-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/20/2022] [Indexed: 02/08/2023] Open
Abstract
Hexokinase 2 (HK2), which catalyzes the first committed step in glucose metabolism, is induced in cancer cells. HK2's role in tumorigenesis has been attributed to its glucose kinase activity. Here, we describe a kinase independent HK2 activity, which contributes to metastasis. HK2 binds and sequesters glycogen synthase kinase 3 (GSK3) and acts as a scaffold forming a ternary complex with the regulatory subunit of protein kinase A (PRKAR1a) and GSK3β to facilitate GSK3β phosphorylation and inhibition by PKA. Thus, HK2 functions as an A-kinase anchoring protein (AKAP). Phosphorylation by GSK3β targets proteins for degradation. Consistently, HK2 increases the level and stability of GSK3 targets, MCL1, NRF2, and particularly SNAIL. In addition to GSK3 inhibition, HK2 kinase activity mediates SNAIL glycosylation, which prohibits its phosphorylation by GSK3. Finally, in mouse models of breast cancer metastasis, HK2 deficiency decreases SNAIL protein levels and inhibits SNAIL-mediated epithelial mesenchymal transition and metastasis.
Collapse
|
14
|
Xu J, Liao K, Yang X, Wu C, Wu W, Han S. Using single-cell sequencing technology to detect circulating tumor cells in solid tumors. Mol Cancer 2021; 20:104. [PMID: 34412644 PMCID: PMC8375060 DOI: 10.1186/s12943-021-01392-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/12/2021] [Indexed: 12/30/2022] Open
Abstract
Circulating tumor cells are tumor cells with high vitality and high metastatic potential that invade and shed into the peripheral blood from primary solid tumors or metastatic foci. Due to the heterogeneity of tumors, it is difficult for high-throughput sequencing analysis of tumor tissues to find the genomic characteristics of low-abundance tumor stem cells. Single-cell sequencing of circulating tumor cells avoids interference from tumor heterogeneity by comparing the differences between single-cell genomes, transcriptomes, and epigenetic groups among circulating tumor cells, primary and metastatic tumors, and metastatic lymph nodes in patients' peripheral blood, providing a new perspective for understanding the biological process of tumors. This article describes the identification, biological characteristics, and single-cell genome-wide variation in circulating tumor cells and summarizes the application of single-cell sequencing technology to tumor typing, metastasis analysis, progression detection, and adjuvant therapy.
Collapse
Affiliation(s)
- Jiasheng Xu
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District Zhejiang Province, Huzhou, China.,Department of Vascular Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Kaili Liao
- Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Xi Yang
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District Zhejiang Province, Huzhou, China
| | - Chengfeng Wu
- Department of Vascular Surgery, the Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Wei Wu
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District Zhejiang Province, 313000, Huzhou, China
| | - Shuwen Han
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District Zhejiang Province, Huzhou, China.
| |
Collapse
|
15
|
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
|
16
|
Wang M, Liao J, Tan C, Zhou H, Wang J, Wang K, Li Y, Wu W. Integrated study of miR-215 promoting breast cancer cell apoptosis by targeting RAD54B. J Cell Mol Med 2021; 25:3327-3338. [PMID: 33635591 PMCID: PMC8034472 DOI: 10.1111/jcmm.16402] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are widely distributed in cells and participate in the regulation of the pathophysiological process of many diseases. As an important part of non-coding RNA, miRNAs regulate a variety of molecules and signal pathways in tumour cells. However, the evidence for regulatory mechanisms of specific miRNAs in tumour cells is still lacking. METHODS In this study, we used transcriptomics analysis and integrated a variety of public databases to screen miRNAs that have key regulatory effects on breast cancer (BC). In addition, we used in vitro and in vivo studies and combined clinical samples to verify its regulatory mechanism. RESULTS We found that among the specific miRNAs, miR-215-5p is a key regulator in BC. Compared with normal adjacent tissues, miR-215-5p has a lower expression level in BC tissues. Patients with high expression levels of miR-215-5p have a longer survival time. miR-215-5p can specifically target the 3'UTR region of RAD54B mRNA and down-regulate the expression of RAD54B, thereby inhibiting the proliferation of BC cells and promoting the apoptosis of BC cells. CONCLUSIONS Finally, we found that miR-215-5p can be used as an important biomarker for BC. We have clarified its function and revealed its mechanism of targeting RAD54B mRNA for the first time. This may provide important clues to reveal the deeper molecular regulation mechanism of BC.
Collapse
Affiliation(s)
- Mingyuan Wang
- Department of PathophysiologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
- Department of Gynaecologythe Affiliated Zhuzhou Hospital Xiangya Medical CollegeCentral South UniversityZhuzhouChina
| | - Jingnan Liao
- Institute of Reproductive and Stem Cell EngineeringSchool of Basic Medical ScienceCentral South UniversityChangshaChina
| | - Chang Tan
- Department of Gynaecologythe Affiliated Zhuzhou Hospital Xiangya Medical CollegeCentral South UniversityZhuzhouChina
| | - Hong Zhou
- Department of Gynaecologythe Affiliated Zhuzhou Hospital Xiangya Medical CollegeCentral South UniversityZhuzhouChina
| | - Jinjin Wang
- Department of Gynaecologythe Affiliated Zhuzhou Hospital Xiangya Medical CollegeCentral South UniversityZhuzhouChina
| | - Kangkai Wang
- Department of PathophysiologySchool of Basic Medical ScienceCentral South UniversityChangshaChina
- Key Laboratory of Sepsis Translational Medicine of HunanCentral South UniversityChangshaChina
- Department of Laboratory AnimalsHunan Key Laboratory of Animal Models for Human DiseasesXiangya School of MedicineCentral South UniversityChangshaChina
| | - Yiming Li
- Department of Geratic SurgeryXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| | - Wei Wu
- Department of Geratic SurgeryXiangya HospitalCentral South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
17
|
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
|
18
|
Chan S, Smith E, Gao Y, Kwan J, Blum BC, Tilston-Lunel AM, Turcinovic I, Varelas X, Cardamone MD, Monti S, Emili A, Perissi V. Loss of G-Protein Pathway Suppressor 2 Promotes Tumor Growth Through Activation of AKT Signaling. Front Cell Dev Biol 2021; 8:608044. [PMID: 33490071 PMCID: PMC7817781 DOI: 10.3389/fcell.2020.608044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
G Protein Suppressor 2 (GPS2) is a multifunctional protein that exerts important roles in inflammation and metabolism in adipose, liver, and immune cells. GPS2 has recently been identified as a significantly mutated gene in breast cancer and other malignancies and proposed to work as a putative tumor suppressor. However, molecular mechanisms by which GPS2 prevents cancer development and/or progression are largely unknown. Here, we have profiled the phenotypic changes induced by GPS2 depletion in MDA-MB-231 triple negative breast cancer cells and investigated the underlying molecular mechanisms. We found that GPS2-deleted MDA-MB-231 cells exhibited increased proliferative, migratory, and invasive properties in vitro, and conferred greater tumor burden in vivo in an orthotopic xenograft mouse model. Transcriptomic, proteomic and phospho-proteomic profiling of GPS2-deleted MBA-MB-231 revealed a network of altered signals that relate to cell growth and PI3K/AKT signaling. Overlay of GPS2-regulated gene expression with MDA-MB-231 cells modified to express constitutively active AKT showed significant overlap, suggesting that sustained AKT activation is associated with loss of GPS2. Accordingly, we demonstrate that the pro-oncogenic phenotypes associated with GPS2 deletion are rescued by pharmacological inhibition of AKT with MK2206. Collectively, these observations confirm a tumor suppressor role for GPS2 and reveal that loss of GPS2 promotes breast cancer cell proliferation and tumor growth through uncontrolled activation of AKT signaling. Moreover, our study points to GPS2 as a potential biomarker for a subclass of breast cancers that would be responsive to PI3K-class inhibitor drugs.
Collapse
Affiliation(s)
- Stefanie Chan
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Emma Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Yuan Gao
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Benjamin C. Blum
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | | | - Isabella Turcinovic
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Maria Dafne Cardamone
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| | - Stefano Monti
- Division of Computational Biology, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
- Center for Network Systems Biology, Boston University, Boston, MA, United States
| | - Valentina Perissi
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
19
|
Shen Q, Zhu H, Lei Q, Chen L, Yang D, Sui W. MicroRNA‑149‑3p inhibits cell proliferation by targeting AKT2 in oral squamous cell carcinoma. Mol Med Rep 2021; 23:172. [PMID: 33398370 PMCID: PMC7821286 DOI: 10.3892/mmr.2020.11811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRs) exhibit oncogenic or tumor suppressive functions that contribute to the initiation and development of various types of human cancer. miR-149-3p has been reported to serve multiple roles in the regulation of proliferation, apoptosis and metastasis. However, the effects and detailed mechanism of miR-149-3p in oral squamous cell carcinoma (OSCC) remain unclear. In the present study, miR-149-3p mimic, mimic control, miR-149-3p inhibitor and inhibitor control were transiently transfected into Cal27 and SCC-9 cells. The viability, proliferation and apoptosis of OSCC cells were determined using Cell Counting Kit-8, colony formation and Annexin V assays, respectively. The mRNA expression levels of miR-149-3p and AKT2 were determined by reverse transcription-quantitative PCR. The protein expression levels of AKT2, cleaved caspase-3 and cleaved PARP were examined by western blot analysis. The binding of miR-149-3p to the AKT2 3′-untranslated region was evaluated by a dual luciferase reporter assay. In the present study, overexpression of miR-149-3p reduced the viability and proliferation of OSCC cells. By contrast, increased cell viability and proliferation was observed in miR-149-3p-deficient OSCC cells. Dual luciferase reporter assay indicated that miR-149-3p significantly decreased the luciferase activity of the wild-type AKT2 3′-untranslated region. Moreover, overexpression of miR-149-3p downregulated the mRNA and protein expression levels of AKT2, suggesting that miR-149-3p was a negative modulator of AKT2. Restoration of AKT2 efficiently reversed the miR-149-3p-mediated reduction in the proliferative capacity of OSCC cells. In addition, miR-149-3p enhanced the sensitivity of OSCC cells to the chemotherapeutic drug 5-fluorouracil. Taken together, the current findings revealed an inhibitory effect of miR-149-3p on the proliferation of OSCC cells through the post-transcriptional suppression of AKT2, and indicated a potential chemosensitizing function of miR-149-3p for the treatment of patients with OSCC.
Collapse
Affiliation(s)
- Qin Shen
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Hong Zhu
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Qiaoling Lei
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Luyuan Chen
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Dajiang Yang
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| | - Wen Sui
- Department of Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong 518100, P.R. China
| |
Collapse
|
20
|
Hay N. How to inhibit breast cancer and breast cancer metastasis with Akt inhibitors: Lessons learned from studies in mice. JOURNAL OF BREAST CANCER RESEARCH 2021; 1:30-33. [PMID: 35578699 PMCID: PMC9107625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The PI3K/Akt signaling pathway is frequently hyperactivated in different types of breast cancer. In the past two decades, major efforts have been made to develop inhibitors of this pathway to treat cancer patients. However, the most evolutionarily conserved function of this pathway is in cellular and organismal metabolism, which is hijacked by cancer cells. Thus, adverse metabolic consequences are expected when PI3K or Akt is targeted. These metabolic consequences, particularly hyperinsulinemia, could impede the efficacy of treatment. This review summarizes recent genetic studies in mice that could pave the way to efficient breast cancer and breast cancer metastasis treatment with Akt inhibitors.
Collapse
Affiliation(s)
- Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA,Research & Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA,Author for correspondence:
| |
Collapse
|
21
|
Csolle MP, Ooms LM, Papa A, Mitchell CA. PTEN and Other PtdIns(3,4,5)P 3 Lipid Phosphatases in Breast Cancer. Int J Mol Sci 2020; 21:ijms21239189. [PMID: 33276499 PMCID: PMC7730566 DOI: 10.3390/ijms21239189] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 12/31/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is hyperactivated in ~70% of breast cancers. Class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane in response to growth factor stimulation, leading to AKT activation to drive cell proliferation, survival and migration. PTEN negatively regulates PI3K/AKT signalling by dephosphorylating PtdIns(3,4,5)P3 to form PtdIns(4,5)P2. PtdIns(3,4,5)P3 can also be hydrolysed by the inositol polyphosphate 5-phosphatases (5-phosphatases) to produce PtdIns(3,4)P2. Interestingly, while PTEN is a bona fide tumour suppressor and is frequently mutated/lost in breast cancer, 5-phosphatases such as PIPP, SHIP2 and SYNJ2, have demonstrated more diverse roles in regulating mammary tumourigenesis. Reduced PIPP expression is associated with triple negative breast cancers and reduced relapse-free and overall survival. Although PIPP depletion enhances AKT phosphorylation and supports tumour growth, this also inhibits cell migration and metastasis in vivo, in a breast cancer oncogene-driven murine model. Paradoxically, SHIP2 and SYNJ2 are increased in primary breast tumours, which correlates with invasive disease and reduced survival. SHIP2 or SYNJ2 overexpression promotes breast tumourigenesis via AKT-dependent and independent mechanisms. This review will discuss how PTEN, PIPP, SHIP2 and SYNJ2 distinctly regulate multiple functional targets, and the mechanisms by which dysregulation of these distinct phosphoinositide phosphatases differentially affect breast cancer progression.
Collapse
|