1
|
Dai L, An D, Huang J, Xiao M, Li Z, Zhou B, Liu H, Xu J, Chen X, Ruan Y. Ovarian multi-omics analysis reveals key rate-limiting enzymes FASN, SCD5, FADS1, 3BHSD, and STAR as potential targets for regulating kidding traits in goats. Int J Biol Macromol 2024; 282:136737. [PMID: 39433193 DOI: 10.1016/j.ijbiomac.2024.136737] [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: 06/17/2024] [Revised: 08/29/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
The kidding traits of goats are an important index of production. However, the molecular regulatory mechanisms of kidding traits in goats have not been fully elucidated. This study aimed to investigate the molecular regulatory network of kidding traits in goats. Multi-omics revealed the enrichment of 10 signaling pathways, with fatty acid biosynthesis, biosynthesis of unsaturated fatty acids, and steroid hormone biosynthesis pathways being closely related to reproduction. Interestingly, the key rate-limiting enzymes, fatty acid synthase (FASN), stearoyl-CoA desaturase 5 (SCD5), fatty acid desaturase 1 (FADS1), 3β-hydroxysteroid dehydrogenase/isomerase (3BHSD), and steroidogenic acute regulatory protein (STAR) enriched in these pathways regulate changes in reproduction-related metabolites. In interference experiments, it was observed that suppressing these key rate-limiting enzymes inhibited the expression of CYP19A1, ESR2, and FSHR. Furthermore, interference inhibited granulosa cell proliferation, caused cell cycle arrest, and promoted apoptosis. Thus, these results suggest that the specific markers of nanny goats with multiple kids are the key rate-limiting enzymes FASN, SCD5, FADS1, 3BHSD, and STAR. These findings may greatly enhance the understanding of regulatory mechanisms that govern goat parturition.
Collapse
Affiliation(s)
- Lingang Dai
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Dongwei An
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiajin Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Meimei Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Ziyang Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Bo Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Jiali Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang 550025, China; College of Animal Science, Guizhou University, Guiyang 550025, China.
| |
Collapse
|
2
|
Ahmad N, Moton S, Kuttikrishnan S, Prabhu KS, Masoodi T, Ahmad S, Uddin S. Fatty acid synthase: A key driver of ovarian cancer metastasis and a promising therapeutic target. Pathol Res Pract 2024; 260:155465. [PMID: 39018927 DOI: 10.1016/j.prp.2024.155465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Fatty acid synthase (FASN) is a critical enzyme essential for the production of fats in the body. The abnormal expression of FASN is associated with different types of malignancies, including ovarian cancer. FASN plays a crucial role in cell growth and survival as a metabolic oncogene, although the specific processes that cause its dysregulation are still unknown. FASN interacts with signaling pathways linked to the progression of cancer. Pharmacologically inhibiting or inactivating the FASN gene has shown potential in causing the death of cancer cells, offering a possible treatment approach. This review examines the function of FASN in ovarian cancer, namely its level of expression, influence on the advancement of the disease, and its potential as a target for therapeutic interventions.
Collapse
Affiliation(s)
- Nuha Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tariq Masoodi
- Cancer Research Department, Sidra Medicine, Doha, Qatar
| | - Sarfraz Ahmad
- Gynecologic Oncology Program, AdventHealth Cancer Institute, Orlando, FL 32804, USA; Florida State University, College of Medicine, Orlando, FL 32801, USA; University of Central Florida, College of Medicine, Orlando, FL 32827, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory of Animal Research Center, Qatar University, Doha 2713, Qatar; Department of Biosciences, Integral University, Lucknow, Uttar Pradesh 226026, India.
| |
Collapse
|
3
|
Chen Y, Yang J, Wan Y, Li Q, Yang D, Wang Y, Gong J, Bai L, Liu Y, Li X, Wang J. DEAR model in overweight endometrial cancer patients undergoing fertility-sparing treatment: A randomized controlled trial. Gynecol Oncol 2024; 185:148-155. [PMID: 38422947 DOI: 10.1016/j.ygyno.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/13/2024] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE To evaluate the effects of DEAR weight management in overweight patients undergoing fertility-sparing treatment for endometrial cancer or atypical hyperplasia. METHODS Women with endometrial cancer or atypical hyperplasia who received fertility-sparing treatment and had a body mass index of >25 kg/m2 were randomly allocated to the DEAR (DEAR weight management) and control (self weight management) groups. Body morphology and composition, glycolipid metabolism, and tumor outcomes were assessed in both groups before and at 3 and 6 months after intervention. RESULTS Overall, 72 subjects were included (36 in each group). Following intervention, the DEAR group showed significantly lower median body weight (69.45 vs. 78.05), body mass index (26.19 vs. 29.15), lipid accumulation index (29.21 vs. 57.86), body fat mass (24.00 vs. 29.30), visceral fat area (112.5 vs. 133.3), and glycolipid metabolic indices (except high density lipoprotein) than the control group (P < 0.05) and showed a decreasing trend. The test group achieved significantly higher complete remission (88.46% vs. 57.14%; P < 0.05); the time to complete remission did not differ significantly (P > 0.05). CONCLUSIONS DEAR weight management can improve the studied parameters and complete remission rates in this population. REGISTRATION NCT06169449.
Collapse
Affiliation(s)
- YiQian Chen
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China; Beijing Health Vocational College, Beijing, China
| | - JingYing Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China; Peking University School of Nursing, Beijing, China
| | - Yu Wan
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - QingRan Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - DanDan Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - YongLi Wang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - JingJing Gong
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - LianHua Bai
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - YuanYuan Liu
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| | - XiaoDan Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China.
| | - JianLiu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
4
|
Hypomethylated gene RAC3 induces cell proliferation and invasion by increasing FASN expression in endometrial cancer. Int J Biochem Cell Biol 2022; 150:106274. [PMID: 35917927 DOI: 10.1016/j.biocel.2022.106274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Endometrial cancer (EC) is one of the most prevalent gynecological cancers with a 5-year survival rate of 20-60%. Feasible prognostic molecular biomarkers of EC are necessary for accurate prediction of EC prognosis. METHODS RAC3 is a member of the Rho GTPases. Public databases including Gene Expression Profiling Interactive Analysis (GEPIA2), Tumor Immune Estimation Resource (TIMER), LinkedOmics, Search Tool for the Retrieval of Interacting Genes/Proteins (STRING), TISIDB and cBioPortal were employed to analyze the differential expression, clinicopathologic characteristics, functional networks, immune cell infiltrates and genetic alteration of RAC3 in EC patients. RESULTS RAC3 expression was elevated in EC patients analyzed by TIMER and GEPIA. Overexpression of RAC3 was obviously correlated with clinical stage, histological type, histological grade and DNA hypomethylation. Patients with high RAC3 expression displayed poor overall survival. Functional enrichment analysis showed that RAC3 was involved in translational initiation, DNA replication and mRNA processing. RAC3 expression was negatively associated with infiltrating levels of B cells, CD8+ T cells, macrophages and dendritic cells in EC. Experiments in vitro showed that RAC3 was upregulated in EC tissues and cell lines, and RAC3 induced cell proliferation and invasion by increasing fatty acid synthase (FASN) expression. CONCLUSION High expression of RAC3iscorrelated with poor prognosis and low infiltration of immune cells in EC. RAC3 promotes cell proliferation and invasion via FASN. These results demonstrate thatRAC3 functions as an EC oncogene and reveal its underlying mechanism in EC progression, suggesting that RAC3 may serve as a potential therapeutic target in EC.
Collapse
|
5
|
Mozihim AK, Chung I, Said NABM, Jamil AHA. Reprogramming of Fatty Acid Metabolism in Gynaecological Cancers: Is There a Role for Oestradiol? Metabolites 2022; 12:metabo12040350. [PMID: 35448537 PMCID: PMC9031151 DOI: 10.3390/metabo12040350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Gynaecological cancers are among the leading causes of cancer-related death among women worldwide. Cancer cells undergo metabolic reprogramming to sustain the production of energy and macromolecules required for cell growth, division and survival. Emerging evidence has provided significant insights into the integral role of fatty acids on tumourigenesis, but the metabolic role of high endogenous oestrogen levels and increased gynaecological cancer risks, notably in obesity, is less understood. This is becoming a renewed research interest, given the recently established association between obesity and incidence of many gynaecological cancers, including breast, ovarian, cervical and endometrial cancers. This review article, hence, comprehensively discusses how FA metabolism is altered in these gynaecological cancers, highlighting the emerging role of oestradiol on the actions of key regulatory enzymes of lipid metabolism, either directly through its classical ER pathways, or indirectly via the IGIFR pathway. Given the dramatic rise in obesity and parallel increase in the prevalence of gynaecological cancers among premenopausal women, further clarifications of the complex mechanisms underpinning gynaecological cancers are needed to inform future prevention efforts. Hence, in our review, we also highlight opportunities where metabolic dependencies can be exploited as viable therapeutic targets for these hormone-responsive cancers.
Collapse
Affiliation(s)
- Azilleo Kristo Mozihim
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nur Akmarina B. M. Said
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, Kuala Lumpur 50603, Malaysia; (A.K.M.); (N.A.B.M.S.)
- Correspondence: ; Tel.: +60-3-7967-4909
| |
Collapse
|
6
|
Dai M, Yang B, Chen J, Liu F, Zhou Y, Zhou Y, Xu Q, Jiang S, Zhao S, Li X, Zhou X, Yang Q, Li J, Wang Y, Zhang Z, Teng Y. Nuclear-translocation of ACLY induced by obesity-related factors enhances pyrimidine metabolism through regulating histone acetylation in endometrial cancer. Cancer Lett 2021; 513:36-49. [PMID: 33991616 DOI: 10.1016/j.canlet.2021.04.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
Endometrial cancer (EC) is becoming one of the most common gynecologic malignancies. Lipid metabolism is a hallmark feature of cancers. The molecular mechanisms underlying lipid metabolism in EC remain unclear. In this study, we revealed that many lipid metabolism-related genes were aberrantly expressed in endometrial cancer tissues, especially ACLY. Upregulated ACLY promoted EC cell proliferation and colony formation, and attenuated apoptosis. Mechanistically, cotreatment with obesity-related factors (estradiol, insulin and leptin) promoted nuclear translocation of ACLY through Akt-mediated phosphorylation of ACLY at Ser455. Nuclear-localized ACLY increased histone acetylation levels, thus resulting in upregulation of pyrimidine metabolism genes, such as DHODH. Moreover, STAT3 altered the ACLY expression at the transcriptional level via directly binding to its promoter region. In conclusion, our findings clarify the roles and mechanisms of ACLY in endometrial cancer and ACLY could link obesity risk factors to the regulation of histone acetylation. We believe that novel therapeutic strategies for EC can be designed by targeting the ACLY axis.
Collapse
Affiliation(s)
- Miao Dai
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China; Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Bikang Yang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China; Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, PR China
| | - Jing Chen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China; Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, PR China
| | - Fei Liu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Yanjie Zhou
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Qinyang Xu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China
| | - Shuheng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200240, Shanghai, PR China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xinchun Li
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Xuan Zhou
- Department of Gynecologic Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Qin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200240, Shanghai, PR China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200240, Shanghai, PR China
| | - Yahui Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200240, Shanghai, PR China
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200240, Shanghai, PR China.
| | - Yincheng Teng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, PR China; Department of Obstetrics and Gynecology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, PR China.
| |
Collapse
|
7
|
Wang Q, Hu R, Li W, Tai Y, Gu W, Das BC, Yang F, Ji J, Wang C, Zhou J. BF175 inhibits endometrial carcinoma through SREBP-regulated metabolic pathways in vitro. Mol Cell Endocrinol 2021; 523:111135. [PMID: 33359761 DOI: 10.1016/j.mce.2020.111135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Elevated lipogenesis is an important metabolic hallmark of rapidly proliferating tumor such as endometrial carcinoma (EC). The sterol regulatory element-binding protein 1 (SREBP1) is a master regulator of lipogenesis and involved in EC proliferation. BF175 is a novel chemical inhibitor of SREBP pathway, and has shown potent anti-lipogenic effects. However, the effect of BF175 on EC cells are yet to be determined. In the present study, we found that BF175 decreased cell viability, colony formation and migratory capacity, inducing autophagy and mitochondrial related apoptosis in EC cell line AN3CA. Z-VAD-FMK partially attenuated the effect of BF175 on AN3CA. In addition, BF175 significantly downregulated SREBPs and their downstream genes. The levels of free fatty acids and total cholesterol were also inhibited. Microarray analysis suggested BF175 treatment obviously affected lipid metabolic pathways in EC. Taken together, we validated BF175 exhibited anti-tumor activity by targeting SREBP-dependent lipogenesis and inducing apoptosis which mitochondrial pathway involved in, suggesting that it's potential as a novel therapeutic reagent for EC.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ruofan Hu
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihua Li
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yanhong Tai
- Department of Pathology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weiting Gu
- Departments of Cancer Biology, Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bhaskar C Das
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fajun Yang
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junyuan Ji
- Department of Molecular and Cellular Medicine, Colleage of Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Chenguang Wang
- Departments of Cancer Biology, Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jie Zhou
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
8
|
Fatty Acid Synthase: An Emerging Target in Cancer. Molecules 2020; 25:molecules25173935. [PMID: 32872164 PMCID: PMC7504791 DOI: 10.3390/molecules25173935] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/22/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
In recent years, lipid metabolism has garnered significant attention as it provides the necessary building blocks required to sustain tumor growth and serves as an alternative fuel source for ATP generation. Fatty acid synthase (FASN) functions as a central regulator of lipid metabolism and plays a critical role in the growth and survival of tumors with lipogenic phenotypes. Accumulating evidence has shown that it is capable of rewiring tumor cells for greater energy flexibility to attain their high energy requirements. This multi-enzyme protein is capable of modulating the function of subcellular organelles for optimal function under different conditions. Apart from lipid metabolism, FASN has functional roles in other cellular processes such as glycolysis and amino acid metabolism. These pivotal roles of FASN in lipid metabolism make it an attractive target in the clinic with several new inhibitors currently being tested in early clinical trials. This article aims to present the current evidence on the emergence of FASN as a target in human malignancies.
Collapse
|
9
|
Evaluation of the antiviral activity of orlistat (tetrahydrolipstatin) against dengue virus, Japanese encephalitis virus, Zika virus and chikungunya virus. Sci Rep 2020; 10:1499. [PMID: 32001767 PMCID: PMC6992670 DOI: 10.1038/s41598-020-58468-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 01/15/2020] [Indexed: 11/20/2022] Open
Abstract
Many mosquito transmitted viruses of the genera Alphavirus and Flavivirus are human pathogens of significant concern, and there is currently no specific antiviral for any member of these two genera. This study sought to investigate the broad utility of orlistat (tetrahydrolipstatin) in reducing virus infection for several mosquito borne viruses including flaviviruses (dengue virus (DENV; nine isolates analyzed), Japanese encephalitis virus (JEV; one isolate analyzed) and Zika virus (ZIKV; 2 isolates analyzed)) as well as an alphavirus (chikungunya virus; CHIKV; 2 isolates analyzed). Three different treatment regimens were evaluated, namely pre-treatment (only), post-treatment (only) and pre- and post-treatment, and three factors were evaluated, namely level of infection, virus titer and genome copy number. Results showed that all three treatment modalities were able to significantly reduce virus titer for all viruses investigated, with the exception of three isolates of DENV in the pre-treatment only regimen. Pre- and post-treatment was more effective in reducing the level of infection and genome copy number of all viruses investigated than either pre-treatment or post-treatment alone. Collectively, these results suggest orlistat has potential as a broad-spectrum agent against multiple mosquito transmitted viruses.
Collapse
|
10
|
Carvalho TM, Cardoso HJ, Figueira MI, Vaz CV, Socorro S. The peculiarities of cancer cell metabolism: A route to metastasization and a target for therapy. Eur J Med Chem 2019; 171:343-363. [PMID: 30928707 DOI: 10.1016/j.ejmech.2019.03.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
The last decade has witnessed the peculiarities of metabolic reprogramming in tumour onset and progression, and their relevance in cancer therapy. Also, it has been indicated that the metastatic process may depend on the metabolic rewiring and adaptation of cancer cells to the pressure of tumour microenvironment and limiting nutrient availability. The present review gatherers the existent knowledge on the influence of tumour microenvironment and metabolic routes driving metastasis. A focus will be given to glycolysis, fatty acid metabolism, glutaminolysis, and amino acid handling. In addition, the role of metabolic waste driving metastasization will be explored. Finally, we discuss the status of cancer treatment approaches targeting metabolism. This knowledge revision will highlight the critical metabolic targets in metastasis and the chemicals already used in preclinical studies and clinical trials, providing clues that would be further exploited in medicinal chemistry research.
Collapse
Affiliation(s)
- Tiago Ma Carvalho
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Henrique J Cardoso
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Marília I Figueira
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia V Vaz
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
11
|
Xia L, Wang H, Cai S, Su X, Shen J, Meng Q, Chen Y, Li L, Yan J, Zhang C, Xu M. Integrated Analysis of a Competing Endogenous RNA Network Revealing a Prognostic Signature for Cervical Cancer. Front Oncol 2018; 8:368. [PMID: 30237984 PMCID: PMC6135876 DOI: 10.3389/fonc.2018.00368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/20/2018] [Indexed: 12/29/2022] Open
Abstract
Given the high morbidity and the trend of younger individuals being affected observed in cervical cancer, it is important to identify sensitive and effective biomarkers for predicting the survival outcome of patients. Based on data from 307 cervical cancer cases acquired from The Cancer Genome Atlas portal, 1920 differentially expressed mRNAs, 70 microRNAs(miRNAs), and 493 long non-coding(lncRNAs) were screened by comparing cervical cancer tissues with paracancerous tissues. A competing endogenous (ceRNA) network containing 50 lncRNAs, 16 miRNAs, and 81 mRNAs was constructed. Eighteen RNAs, comprising 13 mRNAs, 2 miRNAs, and 3 lncRNAs, were identified as significant prognostic factors by univariate Cox proportional hazards regression. ETS-related gene and fatty acid synthase signatures were discovered using a multivariate Cox regression model built to identify independent prognostic factors in cervical cancer patients. Receiver operating characteristic (ROC) analysis was used to determine the optimal cut-off value for distinguishing the risk level of cervical cancer patients. High-risk patients exhibited a poorer prognosis than low-risk patients did. This study focused on ceRNA networks to provide a novel perspective and insight into cervical cancer and suggested that the identified signature can serve as an independent prognostic biomarker in cervical cancer.
Collapse
Affiliation(s)
- Leilei Xia
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Han Wang
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Shengyun Cai
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoling Su
- Department of Obstetrics and Gynecology, No. 455 Hospital, Shanghai, China
| | - Jizi Shen
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qi Meng
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yu Chen
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Li
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiuqiong Yan
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Caihong Zhang
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Mingjuan Xu
- Department of Obstetrics and Gynecology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
12
|
Krencz I, Sebestyen A, Papay J, Jeney A, Hujber Z, Burger CD, Keller CA, Khoor A. In situ analysis of mTORC1/2 and cellular metabolism-related proteins in human Lymphangioleiomyomatosis. Hum Pathol 2018; 79:199-207. [PMID: 29885404 DOI: 10.1016/j.humpath.2018.05.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 10/14/2022]
Abstract
Lymphangioleiomyomatosis (LAM) is a rare progressive cystic lung disease with features of a low-grade neoplasm. It is primarily caused by mutations in TSC1 or TSC2 genes. Sirolimus, an inhibitor of mTOR complex 1 (mTORC1), slows down disease progression in some, but not all patients. Hitherto, other potential therapeutic targets such as mTOR complex 2 (mTORC2) and various metabolic pathways have not been investigated in human LAM tissues. The aim of this study was to assess activities of mTORC1, mTORC2 and various metabolic pathways in human LAM tissues through analysis of protein expression. Immunohistochemical analysis of p-S6 (mTORC1 downstream protein), Rictor (mTORC2 scaffold protein) as well as GLUT1, GAPDH, ATPB, GLS, MCT1, ACSS2 and CPT1A (metabolic pathway markers) were performed on lung tissue from 11 patients with sporadic LAM. Immunoreactivity was assessed in LAM cells with bronchial smooth muscle cells as controls. Expression of p-S6, Rictor, GAPDH, GLS, MCT1, ACSS2 and CPT1A was significantly higher in LAM cells than in bronchial smooth muscle cells (P<.01). No significant differences were found between LAM cells and normal bronchial smooth muscle cells in GLUT1 and ATPB expression. The results are uniquely derived from human tissue and indicate that, in addition to mTORC1, mTORC2 may also play an important role in the pathobiology of LAM. Furthermore, glutaminolysis, acetate utilization and fatty acid β-oxidation appear to be the preferred bioenergetic pathways in LAM cells. mTORC2 and these preferred bioenergetic pathways appear worthy of further study as they may represent possible therapeutic targets in the treatment of LAM.
Collapse
Affiliation(s)
- Ildiko Krencz
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Anna Sebestyen
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Judit Papay
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Andras Jeney
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Zoltan Hujber
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
| | - Charles D Burger
- Division of Pulmonary, Allergy and Sleep Medicine, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Cesar A Keller
- Division of Pulmonary, Allergy and Sleep Medicine, Mayo Clinic, Jacksonville, FL 32224, United States; Division of Transplant Medicine, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Andras Khoor
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Jacksonville, FL 32224, United States.
| |
Collapse
|
13
|
Effects of Fatty Acid Synthase Inhibition by Orlistat on Proliferation of Endometrial Cancer Cell Lines. Target Oncol 2017; 11:763-769. [PMID: 27188391 DOI: 10.1007/s11523-016-0442-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Fatty acid synthase (FAS) is a key lipogenic enzyme that is highly expressed in endometrial cancer. Orlistat is a weight loss medication that has been shown to be a potent inhibitor of FAS. The goal of this study was to evaluate the anti-tumorigenic potential of orlistat in endometrial cancer cell lines. METHODS The endometrial cancer cell lines ECC-1 and KLE were used. Cell proliferation was assessed by MTT assay after treatment with orlistat. Cell cycle progression was evaluated by Cellometer and apoptosis was assessed using the Annexin V assay. Reactive oxygen species (ROS) was measured using the DCFH-DA assay. Western immunoblotting was performed to determine changes in FAS, cellular stress, cell cycle progression, and the AMPK/mTOR pathways. RESULTS Orlistat inhibited cell proliferation by 61 % in ECC-1 cells and 57 % in KLE cells at a dose of 500 μM. Treatment with orlistat at this concentration resulted in G1 arrest (p < 0.05) but did not affect apoptosis. Orlistat increased ROS and induced the expression of BIP (1.28-fold in ECC-1 compared to control, p < 0.05; 1.92-fold in KLE, p < 0.05) and PERK (2.25-fold in ECC-1, 1.4-fold in KLE, p < 0.05). Western immunoblot analysis demonstrated that orlistat decreased expression of important proteins in fatty acid metabolism including FAS (67 % in ECC-1, 15 % in KLE), acetyl-CoA carboxylase (40 % in ECC-1, 35 % in KLE), and carnitine palmitoyltransferase 1A (CPT1A) (65 % in ECC-1, 25 % in KLE) in a dose-dependent manner. In addition, orlistat at a dose of 500 μM increased expression of phosphorylated-AMPK (1.9-fold in ECC-1, p < 0.01; 1.5-fold in KLE, p < 0.05) and decreased expression of phosphorylated-Akt (25 % in ECC-1, p < 0.05; 37 % in KLE, p < 0.05) and phosphorylated-S6 (68 % in ECC-1, 56 % in KLE). CONCLUSIONS Orlistat inhibits cell growth in endometrial cancer cell lines through inhibition of fatty acid metabolism, induction of cell cycle G1 arrest, activation of AMPK and inhibition of the mTOR pathway. Given that patients with endometrial cancer have high rates of obesity, orlistat should be further investigated as a novel strategy for endometrial cancer treatment.
Collapse
|
14
|
Nakayama K, Rahman MT, Rahman M, Nakamura K, Ishikawa M, Katagiri H, Sato E, Ishibashi T, Iida K, Ishikawa N, Kyo S. CCNE1 amplification is associated with aggressive potential in endometrioid endometrial carcinomas. Int J Oncol 2015; 48:506-16. [PMID: 26647729 PMCID: PMC4725452 DOI: 10.3892/ijo.2015.3268] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 11/08/2015] [Indexed: 12/16/2022] Open
Abstract
The clinicopathological significance of amplification was investigated of the gene encoding cyclin E (CCNE1) and we assessed whether CCNE1 was a potential target in endometrioid endometrial carcinomas. CCNE1 amplification and CCNE1 or F-box and WD repeat domain-containing 7 (FBXW7) expression in endometrial endometrioid carcinoma was assessed by immunohistochemistry and fluorescence in situ hybridization. CCNE1 knockdown by small interfering RNA (siRNA) was used to assess the CCNE1 function. The results showed that CCNE1 amplification was present in 9 (8.3%) of 108 endometrial carcinomas. CCNE1 amplification was correlated with high histological grade (Grade 3; P=0.0087) and lymphovascular space invasion (P=0.0258). No significant association was observed between CCNE1 amplification and FIGO stage (P=0.851), lymph node metastasis (P=0.078), body mass index (P=0.265), deep myometrial invasion (P=0.256), menopausal status (P=0.289) or patient age (P=0.0817). CCNE1 amplification was significantly correlated with shorter progression-free and overall survival (P=0.0081 and 0.0073, respectively). CCNE1 protein expression or loss of FBXW7 expression in endometrial endometrioid carcinoma tended to be correlated with shorter progression-free and overall survival; however, this difference was not statistically significant. Multivariate analysis showed that CCNE1 amplification was an independent prognostic factor for overall survival but not for progression-free survival (P=0.0454 and 0.2175, respectively). Profound growth inhibition was observed in siRNA-transfected cancer cells with endogenous CCNE1 overexpression compared with that in cancer cells having low CCNE1 expression. CCNE1 amplification was independent of p53, HER2, MLH1 and ARID1A expression but dependent on PTEN expression in endometrial carcinomas. These findings indicated that CCNE1 amplification was critical for the survival of endometrial endometrioid carcinomas. Furthermore, the effects of CCNE1 knockdown were dependent on the CCNE1 expression status, suggesting that CCNE1-targeted therapy may be beneficial for patients with endometrial endometrioid carcinoma having CCNE1 amplification.
Collapse
Affiliation(s)
- Kentaro Nakayama
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Mohammed Tanjimur Rahman
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Munmun Rahman
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Kohei Nakamura
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Hiroshi Katagiri
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Emi Sato
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Tomoka Ishibashi
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Kouji Iida
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Noriyuki Ishikawa
- Department of Organ Pathology, Shimane University School of Medicine, Izumo 6938501, Japan
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Shimane University School of Medicine, Izumo 6938501, Japan
| |
Collapse
|
15
|
Lawrenson K, Mhawech-Fauceglia P, Worthington J, Spindler TJ, O'Brien D, Lee JM, Spain G, Sharifian M, Wang G, Darcy KM, Pejovic T, Sowter H, Timms JF, Gayther SA. Identification of novel candidate biomarkers of epithelial ovarian cancer by profiling the secretomes of three-dimensional genetic models of ovarian carcinogenesis. Int J Cancer 2015; 137:1806-17. [PMID: 25204737 DOI: 10.1002/ijc.29197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/23/2014] [Accepted: 07/28/2014] [Indexed: 12/27/2022]
Abstract
Epithelial ovarian cancer (EOC) is still considered the most lethal gynecological malignancy and improved early detection of ovarian cancer is crucial to improving patient prognoses. To address this need, we tested whether candidate EOC biomarkers can be identified using three-dimensional (3D) in vitro models. We quantified changes in the abundance of secreted proteins in a 3D genetic model of early-stage EOC, generated by expressing CMYC and KRAS(G) (12) (V) in TERT-immortalized normal ovarian epithelial cells. Cellular proteins were labeled in live cells using stable isotopic amino acid analogues, and secreted proteins identified and quantified using liquid chromatography-tandem mass spectrometry. Thirty-seven and 55 proteins were differentially expressed by CMYC and CMYC+KRAS(G) (12) (V) expressing cells respectively (p < 0.05; >2-fold). We evaluated expression of the top candidate biomarkers in ∼210 primary EOCs: CHI3L1 and FKBP4 are both expressed by >96% of primary EOCs, and FASN and API5 are expressed by 86 and 75% of cases. High expression of CHI3L1 and FKBP4 was associated with worse patient survival (p = 0.042 and p = 0.002, respectively). Expression of LGALS3BP was positively associated with recurrence (p = 0.0001) and suboptimal debulking (p = 0.018) suggesting that these proteins may be novel prognostic biomarkers. Furthermore, within early stage tumours (I/II), high expression of API5, CHI3L1 and FASN was associated with high tumour grade (p = 3 × 10(-4) , p = 0.016, p = 0.010, respectively). We show in vitro cell biology models of early-stage cancer development can be used to identify novel candidate biomarkers for disease, and report the identification of proteins that represent novel potential candidate diagnostic and prognostic biomarkers for this highly lethal disease.
Collapse
Affiliation(s)
- Kate Lawrenson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Paulette Mhawech-Fauceglia
- Departments of Medicine and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Jenny Worthington
- Cancer Proteomics Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Tassja J Spindler
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Darragh O'Brien
- Cancer Proteomics Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Janet M Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Georgia Spain
- Cancer Proteomics Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Maryam Sharifian
- Departments of Medicine and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Guisong Wang
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA
| | - Kathleen M Darcy
- Women's Health Integrated Research Center at Inova Health System, Gynecologic Cancer Center of Excellence, Annandale, VA
| | - Tanja Pejovic
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR
| | - Heidi Sowter
- Biological and Forensic Science Department, University of Derby, Derby, United Kingdom
| | - John F Timms
- Cancer Proteomics Group, Institute for Women's Health, University College London, London, United Kingdom
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
16
|
Wang C, Ma J, Zhang N, Yang Q, Jin Y, Wang Y. The acetyl-CoA carboxylase enzyme: a target for cancer therapy? Expert Rev Anticancer Ther 2015; 15:667-76. [PMID: 25979092 DOI: 10.1586/14737140.2015.1038246] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
As a rate-limiting enzyme, the acetyl-CoA carboxylase (ACC) is essential for fatty acid synthesis. Traditionally, the ACC has been a target of metabolic syndrome and obesity. Recent research has demonstrated that malignant tumors have a high energy flow, thus having a great ability to synthesize fatty acids. ACCs are occasionally found to be overexpressed in cancer cells, and using chemical or RNA interference to inhibit ACC can lead to cancer cell cycle arrest and apoptosis. This suggests that ACC and relative fatty acids may be critical for the survival of cancer cells. In this review, we summarize the role of ACC in tumor development. We also discuss the signaling pathways possibly affected by ACC, which may give insight into future research for cancer therapy.
Collapse
Affiliation(s)
- Chao Wang
- Department of OB&GYN, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | | | | | | | | | | |
Collapse
|
17
|
Ammer E, Nietzsche S, Rien C, Kühnl A, Mader T, Heller R, Sauerbrei A, Henke A. The anti-obesity drug orlistat reveals anti-viral activity. Med Microbiol Immunol 2015; 204:635-45. [DOI: 10.1007/s00430-015-0391-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/06/2015] [Indexed: 12/28/2022]
|
18
|
Elis S, Desmarchais A, Maillard V, Uzbekova S, Monget P, Dupont J. Cell proliferation and progesterone synthesis depend on lipid metabolism in bovine granulosa cells. Theriogenology 2014; 83:840-53. [PMID: 25583222 DOI: 10.1016/j.theriogenology.2014.11.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 12/31/2022]
Abstract
In dairy cows, lipids are essential to support energy supplies for all biological functions, especially during early lactation. Lipid metabolism is crucial for sustaining proper reproductive function. Alteration of lipid metabolism impacts follicular development and affects oocyte developmental competence. Indeed, nonesterified fatty acids are able to decrease granulosa cell (GC) proliferation and affect estradiol synthesis, thus potentially affecting follicular growth and viability. The objective of this study was to assess the impact of lipid metabolism on bovine GCs, through the use of the lipid metabolism inhibitors etomoxir, an inhibitor of fatty acid (FA) oxidation through inhibition of carnitine palmitoyl transferase 1 (CPT1), and C75, an inhibitor of FA synthesis through inhibition of fatty acid synthase. We showed that etomoxir and C75 significantly inhibited DNA synthesis in vitro; C75 also significantly decreased progesterone synthesis. Both inhibitors significantly reduced AMPK (5' adenosine monophosphate-activated protein kinase) and acetyl-CoA carboxylase phosphorylation. Etomoxir also affected the AKT (protein kinase B) signaling pathway. Combined, these data suggest that both FA oxidation and synthesis are important for the bovine GCs to express a proliferative and steroidogenic phenotype and, thus, for sustaining follicular growth. Despite these findings, it is important to note that the changes caused by the inhibitors of FA metabolism on GCs in vitro are globally mild, suggesting that lipid metabolism is not as critical in GCs as was observed in the oocyte-cumulus complex. Further studies are needed to investigate the detailed mechanisms by which lipid metabolism interacts with GC functions.
Collapse
Affiliation(s)
- Sebastien Elis
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France.
| | - Alice Desmarchais
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Virginie Maillard
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Svetlana Uzbekova
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Philippe Monget
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Joëlle Dupont
- INRA, UMR 85 Physiologie de la Reproduction et des Comportements, Nouzilly, France; CNRS, UMR 7247 Physiologie de la Reproduction et des Comportements, Nouzilly, France; Université François Rabelais de Tours, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France; IFCE, UMR Physiologie de la Reproduction et des Comportements, Nouzilly, France
| |
Collapse
|
19
|
Zhou Y, Zhu LB, Peng AF, Wang TF, Long XH, Gao S, Zhou RP, Liu ZL. LY294002 inhibits the malignant phenotype of osteosarcoma cells by modulating the phosphatidylinositol 3‑kinase/Akt/fatty acid synthase signaling pathway in vitro. Mol Med Rep 2014; 11:1352-7. [PMID: 25351625 DOI: 10.3892/mmr.2014.2787] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence suggests that fatty acid synthase (FASN) is crucial in the carcinogenesis of various types of tumor. In addition, the phosphatidylinositol 3‑kinase (PI3K)/Akt signaling pathway, which is closely associated with cellular metabolism, affects cancer biology. However, whether the malignant phenotype of osteosarcoma (OS) cells is regulated by the PI3K/Akt/FASN signaling pathway and how the PI3K family specific inhibitor, 2‑(4‑morpholinyl)‑8‑phenyl‑chromone (LY294002) affects the malignant phenotype of OS cells remains to be elucidated. In the present study, U2‑OS and MG‑63 cells were treated with LY294002 and subsequently western blot analysis was used to examine Akt, p‑Akt and FASN protein expression. Additionally, FASN mRNA was detected by reverse transcription quantitative polymerase chain reaction. MTT and fluorescence‑activated cell sorting assays were used to assess proliferation and apoptosis. Migration and invasion were investigated using wound healing and transwell invasion assays. The results demonstrated that LY294002 suppressed the PI3K/Akt/FASN signaling pathway. However, the malignant phenotypes of OS cells mentioned above were significantly inhibited. The present results indicated that LY294002 inhibits the malignant phenotype of OS cells via modulation of the PI3K/Akt/FASN signaling pathway in vitro and may be a new therapeutic strategy for the management of OS.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Liang Bo Zhu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Ai Fen Peng
- College of Humanities, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| | - Tao Fang Wang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Hua Long
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Song Gao
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Rong Ping Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi Li Liu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
20
|
Yan C, Wei H, Minjuan Z, Yan X, Jingyue Y, Wenchao L, Sheng H. The mTOR inhibitor rapamycin synergizes with a fatty acid synthase inhibitor to induce cytotoxicity in ER/HER2-positive breast cancer cells. PLoS One 2014; 9:e97697. [PMID: 24866893 PMCID: PMC4035285 DOI: 10.1371/journal.pone.0097697] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/23/2014] [Indexed: 12/20/2022] Open
Abstract
Patients with ER/HER2-positive breast cancer have a poor prognosis and are less responsive to selective estrogen receptor modulators; this is presumably due to the crosstalk between ER and HER2. Fatty acid synthase (FASN) is essential for the survival and maintenance of the malignant phenotype of breast cancer cells. An intimate relationship exists between FASN, ER and HER2. We hypothesized that FASN may be the downstream effector underlying ER/HER2 crosstalk through the PI3K/AKT/mTOR pathway in ER/HER2-positive breast cancer. The present study implicated the PI3K/AKT/mTOR pathway in the regulation of FASN expression in ER/HER2-positive breast cancer cells and demonstrated that rapamycin, an mTOR inhibitor, inhibited FASN expression. Cerulenin, a FASN inhibitor, synergized with rapamycin to induce apoptosis and inhibit cell migration and tumorigenesis in ER/HER2-positive breast cancer cells. Our findings suggest that inhibiting the mTOR-FASN axis is a promising new strategy for treating ER/HER2-positive breast cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blotting, Western
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cerulenin/pharmacology
- Drug Synergism
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Fatty Acid Synthase, Type I/antagonists & inhibitors
- Fatty Acid Synthase, Type I/genetics
- Fatty Acid Synthase, Type I/metabolism
- Female
- Fluorescent Antibody Technique, Indirect
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice
- Mice, Nude
- Protein Kinase Inhibitors/pharmacology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sirolimus/pharmacology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Chen Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Huang Wei
- Department of Cardiology, Xi'jing hospital, Fourth military medical university, Xi'an, PR China
| | - Zheng Minjuan
- Department of Ultrasound, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Xue Yan
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yang Jingyue
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Liu Wenchao
- Department of Oncology, Xi'jing Hospital, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| | - Han Sheng
- State Key Laboratory of Military Stomatology, Department of Information Center, School of Stomatology, Fourth Military Medical University, Xi'an, PR China
- * E-mail: (LW); (HS)
| |
Collapse
|
21
|
Wang H, Luo QF, Peng AF, Long XH, Wang TF, Liu ZL, Zhang GM, Zhou RP, Gao S, Zhou Y, Chen WZ. Positive feedback regulation between Akt phosphorylation and fatty acid synthase expression in osteosarcoma. Int J Mol Med 2013; 33:633-9. [PMID: 24366211 DOI: 10.3892/ijmm.2013.1602] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/13/2013] [Indexed: 11/06/2022] Open
Abstract
The activation of PI3K/Akt and the overexpression of fatty acid synthase (FASN) are frequently observed in human osteosarcoma (OS). In the present study, in order to investigate the possible association between the phosphorylation of Akt and FASN expression, immunohistochemical staining was conducted on 24 OS specimens from patients with pulmonary metastasis, which revealed a significant positive correlation between phosphorylated Akt (p-Akt) and the expression of FASN (R=0.469, P=0.04). To investigate the association between p-Akt and FASN in vitro, human U2-OS OS cells were treated with FASN-specific RNAi plasmid or LY294002 (an inhibitor of PI3k/Akt). The mRNA levels of Akt and FASN were measured by real-time PCR. Western blot analysis was also performed to detect the protein experession of PI3K, Akt, p-Akt and FASN. The results demonstrated that the PI3K/Akt signaling pathway modulates FASN expression; the inhibition of FASN resulted in the downregulation of p-Akt in the U2-OS cells. Furthermore, the effects induced by the inhibition of the activity of p-Akt or FASN on the malignant phenotype of U2-OS cells were investigated, demonstrating that the malignant phenotype was inhibited by suppressing the activity of PI3K/Akt or FASN in the U2-OS cells. The findings from our study suggest the existence of a positive feedback regulation between Akt phosphorylation and FASN expression and that this loop may play an important role in the malignant phenotype of OS cells.
Collapse
Affiliation(s)
- Heng Wang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Qing Feng Luo
- Department of Pathology, Cancer Hospital of Jiangxi Province, Nanchang, Jiangxi, P.R. China
| | - Ai Fen Peng
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, P.R. China
| | - Xin Hua Long
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Tao Fang Wang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Zhi Li Liu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Guo Mei Zhang
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Rong Ping Zhou
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Song Gao
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Yang Zhou
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Wen Zhao Chen
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|