1
|
Poenaru RC, Milanesi E, Niculae AM, Dobre AM, Vladut C, Ciocîrlan M, Balaban DV, Herlea V, Dobre M, Hinescu ME. Dysregulation of genes involved in the long-chain fatty acid transport in pancreatic ductal adenocarcinoma. World J Gastrointest Oncol 2025; 17:98409. [DOI: 10.4251/wjgo.v17.i1.98409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/17/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is an aggressive lethal malignancy with limited options for treatment and a 5-year survival rate of 11% in the United States. As for other types of tumors, such as colorectal cancer, aberrant de novo lipid synthesis and reprogrammed lipid metabolism have been suggested to be associated with PDAC development and progression.
AIM To identify the possible involvement of lipid metabolism in PDAC by analyzing in tumoral and non-tumoral tissues the expression level of the most relevant genes involved in the long-chain fatty acid (FA) import into cell.
METHODS A gene expression analysis of FASN, CD36, SLC27A1, SLC27A2, SLC27A3, SLC27A4, SLC27A5, ACSL1, and ACSL3 was performed by qRT-PCR in 24 tumoral PDAC tissues and 11 samples from non-tumoral pancreatic tissues obtained via fine needle aspiration or via surgical resection. The genes were considered significantly dysregulated between the groups when the p value was < 0.05 and the fold change (FC) was ≤ 0.5 and ≥ 2.
RESULTS We found that three FA transporters and two long-chain acyl-CoA synthetases genes were significantly upregulated in the PDAC tissue compared to the non-tumoral tissue: SLC27A2 (FC = 5.66; P = 0.033), SLC27A3 (FC = 2.68; P = 0.040), SLC27A4 (FC = 3.13; P = 0.033), ACSL1 (FC = 4.10; P < 0.001), and ACSL3 (FC = 2.67; P = 0.012). We further investigated any possible association between the levels of the analyzed mRNAs and the specific characteristics of the tumors, including the anatomic location, the lymph node involvement, and the presence of metastasis. A significant difference in the expression of SLC27A3 (FC = 3.28; P = 0.040) was found comparing patients with and without lymph nodes involvement with an overexpression of this transcript in 17 patients presenting tumoral cells in the lymph nodes.
CONCLUSION Despite the low number of patients analyzed, these preliminary results seem to be promising. Addressing lipid metabolism through a broad strategy could be a beneficial way to treat this malignancy. Future in vitro and in vivo studies on these genes may offer important insights into the mechanisms linking PDAC with the long-chain FA import pathway.
Collapse
Affiliation(s)
- Radu Cristian Poenaru
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Elena Milanesi
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Radiobiology, Victor Babes National Institute of Pathology, Bucharest 050096, Romania
| | - Andrei Marian Niculae
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Pathology, Victor Babes National Institute of Pathology, Bucharest 050096, Romania
| | - Anastasia-Maria Dobre
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Catalina Vladut
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Gastroenterology, Prof. Dr. Agrippa Ionescu Clinical Emergency Hospital, Bucharest 011356, Romania
| | - Mihai Ciocîrlan
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Gastroenterology, Prof. Dr. Agrippa Ionescu Clinical Emergency Hospital, Bucharest 011356, Romania
| | - Daniel Vasile Balaban
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
| | - Vlad Herlea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Pathology, Fundeni Clinical Institute, Bucharest 022258, Romania
| | - Maria Dobre
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Pathology, Victor Babes National Institute of Pathology, Bucharest 050096, Romania
| | - Mihail Eugen Hinescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Pathology, Victor Babes National Institute of Pathology, Bucharest 050096, Romania
| |
Collapse
|
2
|
Du S, Wang J, Liu M, Liu R, Wang H, Zhang Y, Zhou F, Pei W. APOM Modulates the Glycolysis Process in Liver Cancer Cells by Controlling the Expression and Activity of HK2 via the Notch Pathway. Biochem Genet 2025:10.1007/s10528-024-11013-y. [PMID: 39754657 DOI: 10.1007/s10528-024-11013-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/20/2024] [Indexed: 01/06/2025]
Abstract
The metabolic pathway of aerobic glycolysis in tumor cells has garnered significant attention in tumor research because of its high activation in cancer cells. Previous research conducted by our team has demonstrated that Apolipoprotein M (APOM) exhibits potential as a factor against liver cancer. However, further investigations are needed to elucidate the precise approach and mechanism that are involved in this process. The findings of this study demonstrated that the inhibition of APOM gene expression led to a notable increase in glucose uptake within liver cancer cells, along with increased levels of lactate dehydrogenase A (LDHA) mRNA and protein expression, as well as increased lactate and adenosine triphosphate (ATP) levels (P < 0.05). These alterations in the cellular microenvironment may be associated with a significant increase in the expression level and enzyme activity of the pivotal enzyme hexokinase 2 (HK2) (P < 0.05). Subsequent investigations revealed notable enrichment of the Notch pathway in liver cancer samples exhibiting low expression of the APOM gene. Western blot experiments demonstrated that the inhibition of APOM gene expression triggers the activation of the Notch pathway in liver cancer cells. Furthermore, the administration of a γ-secretase inhibitor (DAPT) successfully mitigated the increase in HK2 levels, glucose uptake, lactate production, and proliferation of liver cancer cells induced by the downregulation of the APOM gene (P < 0.05). In conclusion, diminished APOM expression may facilitate the progression of liver cancer by stimulating the aerobic glycolysis pathway, which is mediated by the Notch signaling pathway.
Collapse
Affiliation(s)
- Shuangqiu Du
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
| | - Jingtong Wang
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
- School of Clinical Medicine, Wannan Medical Collage, Wuhu, 241002, Anhui, P. R. China
| | - Miaomiao Liu
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
| | - Rong Liu
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
| | - Hui Wang
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
| | - Yao Zhang
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China
| | - Fengcang Zhou
- Basic Teaching Department of Morphology Teaching and Research Section, Anhui College of Traditional Chinese Medicine, Wuhu, 241002, Anhui, P. R. China.
| | - Wenjun Pei
- Anhui Province Key Laboratory of Basic Research and Transformation of Age-Related Diseases, Wannan Medical College, Wuhu, 241002, Anhui, P. R. China.
| |
Collapse
|
3
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
4
|
Zhong X, Zhang W, Zhang W, Yu N, Li W, Song X. FASN contributes to ADM resistance of diffuse large B-cell lymphoma by inhibiting ferroptosis via nf-κB/STAT3/GPX4 axis. Cancer Biol Ther 2024; 25:2403197. [PMID: 39345091 PMCID: PMC11445901 DOI: 10.1080/15384047.2024.2403197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 10/01/2024] Open
Abstract
Drug resistance is a critical impediment to efficient therapy of diffuse large B-cell lymphoma (DLBCL) patients. Recent studies have highlighted the association between ferroptosis and drug resistance that has been reported. Fatty acid synthase (FASN) is always related to a poor prognosis. In this study, we investigate the impact of FASN on drug resistance in DLBCL and explore its potential modulation of ferroptosis mechanisms. The clinical correlation of FASN mRNA expression was first analyzed to confirm the role of FASN on drug resistance in DLBCL based on the TCGA database. Next, the impact of FASN on ferroptosis was investigated in vitro and in vivo. Furthermore, a combination of RNA-seq, western blot, luciferase reporter, and ChIP experiments was employed to elucidate the underlying mechanism. The prognosis for patients with DLBCL was worse when FASN was highly expressed, particularly in those undergoing chemotherapy for Adriamycin (ADM). FASN promoted tumor growth and resistance of DLBCL to ADM, both in vitro and in vivo. It is noteworthy that this effect was achieved by inhibiting ferroptosis, since Fer-1 (a ferroptosis inhibitor) treatment significantly recovered the effects of silencing FASN on inhibiting ferroptosis, while Erastin (a ferroptosis inducer) treatment attenuated the impact of overexpressing FASN. Mechanistically, FASN activated NF-κB/STAT3 signaling pathway through phosphorylating the upstream IKKα and IκBα, and the activated STAT3 promoted GPX4 expression by directly binding to GPX4 promoter. FASN inhibits ferroptosis in DLBCL via NF-κB/STAT3/GPX4 signaling pathway, indicating its critical role in mediating ADM resistance of DLBCL.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Cell Line, Tumor
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm
- Fatty Acid Synthase, Type I/metabolism
- Fatty Acid Synthase, Type I/genetics
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Mice, Nude
- NF-kappa B/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Prognosis
- Signal Transduction/drug effects
- STAT3 Transcription Factor/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Xing Zhong
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Weiwei Zhang
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
- Nanchang Medical College, Nanchang, Jiangxi, P. R. China
| | - Weiming Zhang
- Nanchang Medical College, Nanchang, Jiangxi, P. R. China
| | - Nasha Yu
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Wuping Li
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| | - Xiangxiang Song
- Departments of Lymphatic and Hematological Oncology, Jiangxi Cancer Hospital (The Second Affiliated Hospital of Nanchang Medical College), Nanchang, Jiangxi, P. R. China
- JXHC Key Laboratory of Tumor Microenvironment and Immunoregulation (Jiangxi Cancer Hospital), Nanchang, Jiangxi, P. R. China
| |
Collapse
|
5
|
Zhang J, Xu S, Yue L, Lei H, Zhai X. A Collection of Novel Antitumor Agents That Regulate Lipid Metabolism in the Tumor Microenvironment. J Med Chem 2024. [PMID: 39726379 DOI: 10.1021/acs.jmedchem.4c02809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Lipid metabolism disorder is the cause of one of the most significant metabolic changes in tumors. In the process of tumor occurrence and development, tumor cells choose a continuous metabolic adaptation to accommodate the changing environment to the maximum extent possible. In a variety of tumors, the uptake, production, and storage of lipids are generally upregulated. Tumor cells take advantage of lipid metabolism to access basic energy, biofilm components, and signal molecules of the tumor microenvironment required for proliferation, survival, invasion, and metastasis. This Perspective briefly uncovers the main metabolic processes and key factors involved in lipid metabolism reprogramming, mainly related to lipid uptake, de novo synthesis and storage of fatty acids, oxidation of fatty acids, cholesterol synthesis, and related regulatory factors. From a medicinal chemistry perspective, agents against related key targets are reviewed, expecting to pave the way for promising antitumor drugs with prospects for application through lipid metabolism reprogramming.
Collapse
Affiliation(s)
- Jiahao Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Sha Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Lingfeng Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Hongrui Lei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| |
Collapse
|
6
|
Chaturvedi S, Sonawane A. Recapitulating the potential contribution of protein S-palmitoylation in cancer. Cancer Metastasis Rev 2024; 44:20. [PMID: 39725785 DOI: 10.1007/s10555-024-10217-3] [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: 06/10/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
Protein S-palmitoylation is a reversible form of protein lipidation in which the formation of a thioester bond occurs between a cysteine (Cys) residue of a protein and a 16-carbon fatty acid chain. This modification is catalyzed by a family of palmitoyl acyl transferases, the DHHC enzymes, so called because of their Asp-His-His-Cys (DHHC) catalytic motif. Deregulation of DHHC enzymes has been linked to various diseases, including cancer and infections. Cancer, a major cause of global mortality, is characterized by features like uncontrolled cell growth, resistance to cell death, angiogenesis, invasion, and metastasis. Several of these processes are controlled by DHHC-mediated S-palmitoylation of oncogenes or tumor suppressors, including growth factor receptors (e.g., EGFR), kinases (e.g., AKT), and transcription factors (e.g., β-catenin). Dynamic regulation of S-palmitoylation is also governed by protein depalmitoylases. These enzymes balance the cycling of palmitoylation and regulate cellular signaling, cell growth, and its organization. Given the significance of S-palmitoylation in cancer, the DHHCs and protein depalmitoylases are promising targets for cancer therapy. Here we summarize the catalytic mechanisms of DHHC enzymes and depalmitoylases, their role in cancer progression and prevention, as well as the crosstalk of palmitoylation with other post-translational modifications. Additionally, we discuss the methods to detect S-palmitoylation, the limitations of available DHHC-targeting inhibitors, and ongoing research efforts to address these obstacles.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India.
| |
Collapse
|
7
|
Dong H, Chen J, Zhang H, Zhao M, Yue Y, Wang S. Palmitic acid inhibits macrophage-mediated chemotherapy resistance in multiple myeloma via ALOX12 signaling. Int Immunopharmacol 2024; 143:113320. [PMID: 39378653 DOI: 10.1016/j.intimp.2024.113320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
We previously discovered that macrophages (MΦs), especially tumor-associated MΦs (tMΦs), contribute to chemotherapy resistance in multiple myeloma (MM). However, the mechanism underlying MΦ-mediated chemotherapy resistance in MM needs further elucidation, and the identification of factors that preferentially abrogate MΦ-induced inhibition of MM chemotherapy may have important clinical significance. In this study, we showed that the expression of FASN and SCD2, the enzymes that synthesize palmitic acid and convert it to palmitoleic acid, was decreased in tMΦs compared with MΦs. Interestingly, palmitic acid abrogated the MΦ-mediated protection of MM cells from the effects of bortezomib and melphalan in vitro. Combination treatment with palmitic acid and bortezomib or melphalan further inhibited MM tumor growth in vivo. Mechanistically, palmitic acid treatment increased ALOX12 expression in MΦs. ALOX12 inhibition partially abrogated the palmitic acid-induced decrease in MΦ-mediated MM cell survival. Palmitic acid treatment inhibited AMPK signaling in MΦs, and ALOX12 knockdown activated the AMPK signaling pathway in MΦs. AMPK inhibition decreased the MΦ-mediated protection of drug-treated MM cells, and AMPK activation partially abolished the palmitic acid-induced inhibition of MΦ-mediated protection. ALOX12 converts arachidonic acid (AA) to 12-HETE. Moreover, treatment with AA but not 12-HETE partially abrogated the inhibitory effect of palmitic acid on MΦ-mediated MM cell survival in response to bortezomib or melphalan. Overall, we identified palmitic acid as a factor that inhibits MΦ-mediated resistance to bortezomib and melphalan in MM, which may have clinical significance.
Collapse
Affiliation(s)
- He Dong
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jintong Chen
- Department of Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Hua Zhang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Munan Zhao
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Ying Yue
- Department of Gynecological Oncology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Siqing Wang
- Department of Cancer Immunology, The First Hospital of Jilin University, Changchun 130061, China.
| |
Collapse
|
8
|
Mo Y, Han Y, Chen Y, Fu C, Li Q, Liu Z, Xiao M, Xu B. ZDHHC20 mediated S-palmitoylation of fatty acid synthase (FASN) promotes hepatocarcinogenesis. Mol Cancer 2024; 23:274. [PMID: 39696259 DOI: 10.1186/s12943-024-02195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Protein palmitoylation is a reversible fatty acyl modification that undertakes important functions in multiple physiological processes. Dysregulated palmitoylations are frequently associated with the formation of cancer. How palmitoyltransferases for S-palmitoylation are involved in the occurrence and development of hepatocellular carcinoma (HCC) is largely unknown. METHODS Chemical carcinogen diethylnitrosamine (DEN)-induced and DEN combined CCl4 HCC models were used in the zinc finger DHHC-type palmitoyltransferase 20 (ZDHHC20) knockout mice to investigate the role of ZDHHC20 in HCC tumourigenesis. Palmitoylation liquid chromatography-mass spectrometry analysis, acyl-biotin exchange assay, co-immunoprecipitation, ubiquitination assays, protein half-life assays and immunofluorescence microscopy were conducted to explore the downstream regulators and corresponding mechanisms of ZDHHC20 in HCC. RESULTS Knocking out of ZDHHC20 significantly reduced hepatocarcinogenesis induced by chemical agents in the two HCC mouse models in vivo. 97 proteins with 123 cysteine sites were found to be palmitoylated in a ZDHHC20-dependent manner. Among these, fatty acid synthase (FASN) was palmitoylated at cysteines 1471 and 1881 by ZDHHC20. The genetic knockout or pharmacological inhibition of ZDHHC20, as well as the mutation of the critical cysteine sites of FASN (C1471S/C1881S) accelerated the degradation of FASN. Furthermore, ZDHHC20-mediated FASN palmitoylation competed against the ubiquitin-proteasome pathway via the E3 ubiquitin ligase complex SNX8-TRIM28. CONCLUSIONS Our findings demonstrate the critical role of ZDHHC20 in promoting hepatocarcinogenesis, and a mechanism underlying a mutual restricting mode for protein palmitoylation and ubiquitination modifications.
Collapse
Affiliation(s)
- Yaqi Mo
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Yamei Han
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yang Chen
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Radiation Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Chunling Fu
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qing Li
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China
| | - Zhuang Liu
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Mingming Xiao
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Bo Xu
- Center for Intelligent Oncology, Chongqing University Cancer Hospital and Chongqing University School of Medicine, and Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Chongqing, 400030, China.
- Department of Biochemistry and Molecular Biology, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| |
Collapse
|
9
|
Kandasamy T, Sarkar S, Ghosh SS. Harnessing Drug Repurposing to Combat Breast Cancer by Targeting Altered Metabolism and Epithelial-to-Mesenchymal Transition Pathways. ACS Pharmacol Transl Sci 2024; 7:3780-3794. [PMID: 39698277 PMCID: PMC11650739 DOI: 10.1021/acsptsci.4c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Breast cancer remains one of the most prevalent and challenging cancers to treat due to its complexity and heterogenicity. Cellular processes such as metabolic reprogramming and epithelial-to-mesenchymal transition (EMT) contribute to the complexity of breast cancer by driving uncontrolled cell division, metastasis, and resistance to therapies. Strategically targeting these intricate pathways can effectively impede breast cancer progression, thereby revealing significant potential for therapeutic interventions. Among various emerging therapeutic approaches, drug repurposing offers a promising avenue for enhancing clinical outcomes. In recent years, high-throughput screening, QSAR, and network pharmacology have been widely employed to identify promising repurposed drugs. As an outcome, several drugs, such as Metformin, Itraconazole, Pimozide, and Disulfiram, were repurposed to regulate metabolic and EMT pathways. Moreover, strategies such as combination therapy, targeted delivery, and personalized medicine were utilized to enhance the efficacy and specificity of the repurposed drugs. This review focuses on the potential of targeting altered metabolism and EMT in breast cancer through drug repurposing. It also highlights recent advancements in drug screening techniques, associated limitations, and strategies to overcome these challenges.
Collapse
Affiliation(s)
- Thirukumaran Kandasamy
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati-39, Assam India
| | - Shilpi Sarkar
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati-39, Assam India
| | - Siddhartha Sankar Ghosh
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati-39, Assam India
- Centre
for Nanotechnology, Indian Institute of
Technology Guwahati, Guwahati-39, Assam India
| |
Collapse
|
10
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2024; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
11
|
Ju R, Ying Y, Zhou Q, Cao Y. Exploring Genetic Drug Targets in Acne Vulgaris: A Comprehensive Proteome-Wide Mendelian Randomization Study. J Cosmet Dermatol 2024; 23:4223-4229. [PMID: 39297226 PMCID: PMC11626312 DOI: 10.1111/jocd.16567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/20/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Acne vulgaris presents a substantial clinical challenge due to its complex pathophysiology and significant impact on quality of life. Identification of novel therapeutic targets for acne using genetic tools can guide the development of more effective treatments. METHODS Utilizing a dataset comprising 35 559 Icelandic individuals, we performed proteomic analyses to quantify 4709 circulating proteins. We integrated these data with acne-specific genome-wide association studies (GWAS) encompassing 34 422 acne patients and 364 991 controls. Mendelian randomization (MR) analyses employed the TwoSampleMR tool and Summary-data-based Mendelian Randomization (SMR) to estimate the causal effects of identified proteins on acne risk. Colocalization analyses assessed the likelihood of shared genetic etiology between protein levels and acne using the "coloc" R package. RESULTS Our proteome-wide MR analysis initially identified 128 proteins potentially associated with acne risk. Following multiple testing corrections using the Benjamini-Hochberg method, fatty acid synthase (FASN) and tissue inhibitor of metalloproteinases 4 (TIMP4) remained significantly associated with acne risk. FASN exhibited a protective effect against acne (OR = 0.768, 95% CI: 0.676-0.872, p = 4.685E-05), while TIMP4 was associated with an increased risk (OR = 1.169, 95% CI: 1.103-1.241, p = 1.956E-07). Colocalization analysis supported a shared genetic basis for these protein-acne associations, with posterior probabilities indicating strong evidence of shared causal variants. CONCLUSION Our findings highlight the utility of integrative genomic approaches in identifying potential therapeutic targets for acne. FASN and TIMP4, in particular, demonstrate strong potential as targets for therapeutic intervention, pending further validation through clinical research. These results offer a foundation for targeted acne treatment development, aligning with personalized medicine principles.
Collapse
Affiliation(s)
- Ruyi Ju
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhou CityZhejiang ProvinceChina
| | - Yuou Ying
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhou CityZhejiang ProvinceChina
| | - Qiujun Zhou
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhou CityZhejiang ProvinceChina
| | - Yi Cao
- Zhejiang Chinese Medical University and the First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou CityZhejiang ProvinceChina
| |
Collapse
|
12
|
Chang Y, Du R, Xia F, Xu X, Wang H, Chen X. Dysregulation of Fatty Acid Metabolism in Breast Cancer and Its Targeted Therapy. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:825-844. [PMID: 39628960 PMCID: PMC11614585 DOI: 10.2147/bctt.s496322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/13/2024] [Indexed: 12/06/2024]
Abstract
Breast cancer has become the number one cancer worldwide, there are challenges in its prevention, diagnosis and treatment, especially the pathogenesis of triple negative breast cancer has not been clear and the treatment dilemma of metastatic breast cancer. Metabolic reprogramming is currently considered to be one of the hallmarks of cancer, and metabolic alterations in breast cancer, including enhanced glycolysis, tricarboxylic acid cycle activity, glutamine catabolism, and fatty acid biosynthesis, are manifested differently in different breast cancer subtypes and have a complex relationship with tumor growth, metastasis, death, and drug resistance. At present, inhibitors of fatty acid synthesis and oxidation related enzymes have a certain effect in the treatment of breast cancer. In this paper, we review the studies on fatty acid metabolism in breast cancer to better understand the mechanism of fatty acid metabolism in breast cancer pathogenesis and hope to provide new ideas for targeting fatty acid metabolism in the treatment of breast cancer.
Collapse
Affiliation(s)
- Yue Chang
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Rui Du
- Department of Anorectal Surgery, The Third Affiliated Hospital of Anhui Medical University, Hefei First People’s Hospital, Hefei, Anhui, People’s Republic of China
| | - Fan Xia
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Xiuli Xu
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Hongzhi Wang
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| | - Xueran Chen
- Hefei Cancer Hospital of CAS; Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences (CAS), Hefei, Anhui, People’s Republic of China
| |
Collapse
|
13
|
Zhao Y, Liu MJ, Zhang L, Yang Q, Sun QH, Guo JR, Lei XY, He KY, Li JQ, Yang JY, Jian YP, Xu ZX. High mobility group A1 (HMGA1) promotes the tumorigenesis of colorectal cancer by increasing lipid synthesis. Nat Commun 2024; 15:9909. [PMID: 39548107 PMCID: PMC11568219 DOI: 10.1038/s41467-024-54400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to meet the high energy and biosynthetic demands required for their proliferation. High mobility group A1 (HMGA1) is a structural transcription factor and frequently overexpressed in human colorectal cancer (CRC). Here, we show that HMGA1 promotes CRC progression by driving lipid synthesis in a AOM/DSS-induced CRC mouse model. Using conditional knockout (Hmga1△IEC) and knock-in (Hmga1IEC-OE/+) mouse models, we demonstrate that HMGA1 enhances CRC cell proliferation and accelerates tumor development by upregulating fatty acid synthase (FASN). Mechanistically, HMGA1 increases the transcriptional activity of sterol regulatory element-binding protein 1 (SREBP1) on the FASN promoter, leading to increased lipid accumulation in intestinal epithelial cells. Moreover, a high-fat diet exacerbates CRC progression in Hmga1△IEC mice, while pharmacological inhibition of FASN by orlistat reduces tumor growth in Hmga1IEC-OE/+ mice. Our findings suggest that targeting lipid metabolism could offer a promising therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qi Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province, China.
| |
Collapse
|
14
|
Zhou S, Zhang Q, Xu J, Xiang R, Dong X, Zhou X, Liu Z. CAP superfamily proteins in human: a new target for cancer therapy. Med Oncol 2024; 41:306. [PMID: 39499355 DOI: 10.1007/s12032-024-02548-6] [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: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/07/2024]
Abstract
The CAP (Cysteine-rich secretory protein, Antigen 5, and Pathogenesis-related protein 1) superfamily proteins (CAP proteins) are found in all kingdoms of life. The cysteine-rich secreted proteins are prevalent in human organs and tissues and serve as critical signaling molecules within cells, regulating a wide range of biochemical processes in the human body. Due to their involvement in numerous biological processes, CAP proteins have recently attracted significant attention, particularly in the context of tumorigenesis and cancer therapy. This review summarizes the expression patterns and roles of CAP proteins in various cancers. Additionally, it analyzes the mechanisms by which CAP proteins affect cancer cell proliferation and survival, regulate epithelial-mesenchymal transition, influence drug resistance, and regulate epigenetics. The review reveals that CAP proteins play distinct roles in various signaling pathways, such as the MAPK, PI3K-Akt, and p53 pathways, which are crucial for tumor progression. Furthermore, this review summarizes the tumor-inhibiting function of CAP proteins and their potential as cancer biomarkers. These findings suggest that CAP proteins represent a promising new target for innovative cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Shenao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Qianqian Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Jiawei Xu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ruiqi Xiang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Xiaoping Dong
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Xi Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
| | - Zhonghua Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
- Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
15
|
Jia L, Peng J, Chen H, Liu Z, Gong J, Sun N, Zhang Q, Li L. TPTEP1 impedes the reprogramming of fatty acid metabolism in triple negative breast cancer via miR-1343-3p/SIRT3 axis. Int J Biol Macromol 2024; 280:135792. [PMID: 39304052 DOI: 10.1016/j.ijbiomac.2024.135792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Recently, the important role of fatty acid (FA) metabolism in cancers has been highlighted. Sirtuin 3 (SIRT3) is determined as an important regulator in the FA metabolism of cancer cells. We are going to verify whether and how lncRNA transmembrane phosphatase with tensin homology pseudogene 1 (TPTEP1) and SIRT3 may exert certain impact on the FA metabolism in triple-negative breast cancer (TNBC). Firstly, TPTEP1 was verified to be with low expression in TNBC cells. Moreover, down-regulation of TPTEP1 was caused by YY1 transcription factor. Functional assays determined the effects of TPTEP1 on the process of TNBC. The results disclosed that TPTEP1 up-regulation significantly repressed cell proliferation, migration, invasion, EMT and the reprogramming of FA metabolism in TNBC. Mechanism experiments detected the regulatory mechanism between TPTEP1 and SIRT3, which turned out that TPTEP1 positively regulated SIRT3 to affect FOXO3a and inhibit the Wnt/β-catenin pathway via sponging miR-1343-3p. All in all, TPTEP1 functioned as a tumor suppressor to regulate TNBC progression via the miR-1343-3p/SIRT3/FOXO3a/Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Lin Jia
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Junning Peng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Hongying Chen
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Zhenyu Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Jiaxin Gong
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Nan Sun
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| | - Liru Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| |
Collapse
|
16
|
Liang Y, Qi J, Yu D, Wang Z, Li W, Long F, Ning S, Yuan M, Zhong X. Ferulic Acid Alleviates Lipid and Bile Acid Metabolism Disorders by Targeting FASN and CYP7A1 in Iron Overload-Treated Mice. Antioxidants (Basel) 2024; 13:1277. [PMID: 39594419 PMCID: PMC11591460 DOI: 10.3390/antiox13111277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/06/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024] Open
Abstract
Iron overload is a common complication in various chronic liver diseases, including non-alcoholic fatty liver disease (NAFLD). Lipid and bile acid metabolism disorders are regarded as crucial hallmarks of NAFLD. However, effects of iron accumulation on lipid and bile acid metabolism are not well understood. Ferulic acid (FA) can chelate iron and regulate lipid and bile acid metabolism, but its potential to alleviate lipid and bile acid metabolism disorders caused by iron overload remains unclear. Here, in vitro experiments, iron overload induced oxidative stress, apoptosis, genomic instability, and lipid deposition in AML12 cells. FA reduced lipid and bile acid synthesis while increasing fatty acid β-oxidation and bile acid export, as indicated by increased mRNA expression of PPARα, Acox1, Adipoq, Bsep, and Shp, and decreased mRNA expression of Fasn, Acc, and Cyp7a1. In vivo experiments, FA mitigated liver injury in mice caused by iron overload, as indicated by reduced AST and ALT activities, and decreased iron levels in both serum and liver. RNA-seq results showed that differentially expressed genes were enriched in biological processes related to lipid metabolism, lipid biosynthesis, lipid storage, and transport. Furthermore, FA decreased cholesterol and bile acid contents, downregulated lipogenesis protein FASN, and bile acid synthesis protein CYP7A1. In conclusion, FA can protect the liver from lipid and bile acid metabolism disorders caused by iron overload by targeting FASN and CYP7A1. Consequently, FA, as a dietary supplement, can potentially prevent and treat chronic liver diseases related to iron overload by regulating lipid and bile acid metabolism.
Collapse
Affiliation(s)
- Yaxu Liang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Jun Qi
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Dongming Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Zhibo Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Weite Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Fei Long
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Shuai Ning
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Meng Yuan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
| | - Xiang Zhong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.L.); (J.Q.); (D.Y.); (Z.W.); (W.L.); (F.L.); (S.N.); (M.Y.)
- Natural Plant and Animal Health Innovation Institute, NJAU-Cohoo Biotechnology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
17
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
18
|
Shang Z, Arishi AA, Wu C, Lao F, Gilchrist CLM, Moggach SA, Lacey E, Piggott AM, Chooi YH. Self-Resistance Gene-Guided Discovery of the Molecular Basis for Biosynthesis of the Fatty Acid Synthase Inhibitor Cerulenin. Angew Chem Int Ed Engl 2024:e202414941. [PMID: 39363718 DOI: 10.1002/anie.202414941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024]
Abstract
Cerulenin (1) is the first reported natural fatty acid synthase inhibitor and has been intensively researched for its antifungal, anticancer and anti-obesity properties. However, the molecular basis for its biosynthesis has remained a mystery for six decades. Here, we have identified the polyketide biosynthetic gene cluster (cer) responsible for the biosynthesis of 1 from two Sarocladium species using a self-resistance gene mining approach, which we validated via heterologous reconstitution of cer cluster in an Aspergillus nidulans host. Expression of various combinations of cer genes uncovered key pathway intermediates, electrocyclisation products derived from PKS-encoded polyenoic acids, and a suite of 13 new analogues of 1. This enabled us to establish a biosynthetic pathway to 1 that starts with a C12 polyketide precursor containing both E and Z double bonds and involves a complex series of epoxidations, double bond shifts, E/Z isomerisation and epoxide reduction. Using in vitro assays, we further validated the roles of amidotransferase CerD in amidation, and oxidase CerF and reductase CerE in the final two-electron oxidation and enone reduction steps towards 1. These findings expand our understanding of complex tailoring modifications in highly reducing PKS pathways and pave the way for the engineered biosynthesis of cerulenin analogues.
Collapse
Affiliation(s)
- Zhuo Shang
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
- School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, China
| | - Amr A Arishi
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
- Department of Botany and Microbiology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Changzheng Wu
- School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, China
| | - Fangzheng Lao
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
| | - Cameron L M Gilchrist
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
- Present address: School of Biological Sciences, Seoul National University, 08826, Seoul, South Korea
| | - Stephen A Moggach
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
| | - Ernest Lacey
- Microbial Screening Technologies Pty. Ltd., 2164, Smithfield, NSW, Australia
- School of Natural Sciences, Macquarie University, 2109, Sydney, NSW, Australia
| | - Andrew M Piggott
- School of Natural Sciences, Macquarie University, 2109, Sydney, NSW, Australia
| | - Yit-Heng Chooi
- School of Molecular Sciences, The University of Western Australia, 6009, Perth, WA, Australia
| |
Collapse
|
19
|
Liu W, Deng J, Tao XJ, Peng Y, Chen XD, Qu XC, Deng HW, Tan LJ. Aurantio‑obtusin regulates lipogenesis and ferroptosis of liver cancer cells through inhibiting SCD1 and sensitizing RSL3. Int J Oncol 2024; 65:92. [PMID: 39155877 PMCID: PMC11374152 DOI: 10.3892/ijo.2024.5680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Ferroptosis, characterized by iron‑mediated non‑apoptotic cell death and alterations in lipid redox metabolism, has emerged as a critical process implicated in various cellular functions, including cancer. Aurantio‑obtusin (AO), a bioactive compound derived from Cassiae semen (the dried mature seeds of Cassie obtusifolia L. or Cassia toral L.), has anti‑hyperlipidemic and antioxidant properties; however, to the best of our knowledge, the effect of AO on liver cancer cells remains unclear. The Cell Counting Kit‑8, EdU staining and migration assays were employed to assess the anti‑liver cancer activity of AO. Intracellular levels of glutathione peroxidase 4 protein and lipid peroxidation were measured as indicators of ferroptotic status. Immunohistochemical analyses, bioinformatics analyses and western blotting were conducted to evaluate the potential of stearoyl‑CoA desaturase 1 (SCD1) in combination with ferroptosis inducers for the personalized treatment of liver cancer. The present study revealed that AO significantly inhibited the proliferation of liver cancer cells in vitro and in vivo. Mechanistically, AO inhibited AKT/mammalian target of rapamycin (mTOR) signaling, suppressed sterol regulatory element‑binding protein 1 (SREBP1) expression, and downregulated fatty acid synthase expression, thereby inhibiting de novo fatty acid synthesis. Further investigations demonstrated that AO suppressed glutathione peroxidase 4 protein expression through the nuclear factor erythroid 2‑related factor 2/heme oxygenase‑1 pathway, induced ferroptosis in liver cancer cells, and simultaneously inhibited lipogenesis by suppressing SCD1 expression through the AKT/mTOR/SREBP1 pathway. Consequently, this increased the sensitivity of liver cancer cells to the ferroptosis inducer RSL3. Additionally, the enhanced effects of AO and RSL3, which resulted in significant tumor suppression, were confirmed in a xenograft mouse model. In conclusion, the present study demonstrated that AO induced ferroptosis, downregulated the expression of SCD1 and enhanced the sensitivity of liver cancer cells to the ferroptosis inducer RSL3. The synergistic use of AO and a ferroptosis inducer may have promising therapeutic effects in liver cancer cells.
Collapse
Affiliation(s)
- Wen Liu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410000, P.R. China
| | - Xiao-Jun Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Ya Peng
- Department of Pharmacy, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410000, P.R. China
| | - Xiang-Ding Chen
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiao-Chao Qu
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Hong-Wen Deng
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Li-Jun Tan
- Laboratory of Molecular and Statistical Genetics, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
20
|
Paunovic M, Stojanovic A, Pokimica B, Martacic JD, Cvetkovic Z, Ivanovic N, Vucic V. Metabolic Reprogramming of Phospholipid Fatty Acids as a Signature of Lung Cancer Type. Cancers (Basel) 2024; 16:3320. [PMID: 39409945 PMCID: PMC11475191 DOI: 10.3390/cancers16193320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Lung cancer is one of the leading causes of cancer-related mortality. Non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC) differ in aggressiveness, proliferation speed, metastasis propensity, and prognosis. Since tumor cells notably change lipid metabolism, especially phospholipids and fatty acids (FA), this study aimed to identify FA alterations in lung cancer tissues. Methods: Our study included patients with newly diagnosed, histologically confirmed SCLC (n = 27) and NSCLC (n = 37). Samples were collected from both malignant and healthy tissues from each patient, providing they were within subject design. Results: In both NSCLC and SCLC tumor tissues, FA contents were shifted toward pro-inflammatory profiles, with increased levels of some individual n-6 polyunsaturated FA (PUFA), particularly arachidonic acid, and elevated activity of Δ6 desaturase. Compared to healthy counterparts, lower levels of alpha-linolenic acid (18:3n-3) and total saturated FA (SFA) were found in NSCLC, while decreased levels of linoleic acid (18:2n-6) and all individual n-3 FA were found in SCLC tissue in comparison to the healthy tissue control. When mutually compared, SCLC tissue had higher levels of total SFA, especially stearic acid, while higher levels of linoleic acid, total PUFA, and n-3 and n-6 PUFA were detected in NSCLC. Estimated activities of Δ6 desaturase and elongase were higher in SCLC than in NSCLC. Conclusions: Our findings indicate a notable impairment of lipid metabolism in two types of lung cancer tissues. These type-specific alterations may be associated with differences in their progression and also point out different therapeutic targets.
Collapse
Affiliation(s)
- Marija Paunovic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Ana Stojanovic
- Department of Pulmonology, University Hospital Medical Center (UHMC) “Bezanijska kosa”, 11000 Belgrade, Serbia;
| | - Biljana Pokimica
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Jasmina Debeljak Martacic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| | - Zorica Cvetkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.C.); (N.I.)
- Department of Hematology, University Hospital Medical Center Zemun, 11080 Belgrade, Serbia
| | - Nebojsa Ivanovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (Z.C.); (N.I.)
- Department of Surgical Oncology, University Hospital Medical Center (UHMC) “Bezanijska kosa”, 11000 Belgrade, Serbia
| | - Vesna Vucic
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia; (B.P.); (J.D.M.)
| |
Collapse
|
21
|
Zhi L, Li H, Shi B, Yu T, Jia X, Zhang H. Design, synthesis and neuroprotective activity of compound derived from Gastrodia elata Blume and borneol. Front Pharmacol 2024; 15:1437806. [PMID: 39376614 PMCID: PMC11456490 DOI: 10.3389/fphar.2024.1437806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Traditional Chinese medicine Gastrodia elata Blume (GEB) possesses properties that soothe the liver and dispel wind. Its constituents exhibit numerous pharmacological properties, including neuroprotective effects, analgesic properties for headache relief, memory enhancement, and others. Borneol enhances drug absorption by traversing the blood-brain barrier, thereby improving its bioavailability and therapeutic efficacy. The research aimed to design innovative drug molecules and contribute to the beneficial exploration of compound Chinese medicine modernization. Methods This study employed the strategy of "compound Chinese medicine molecular chemistry" to integrate and fuse the effective substances of compound Chinese medicines. An excitotoxic injury model was established by exposing PC12 cells to glutamate. Cell viability was quantitatively evaluated utilizing a colorimetric assay with the CCK-8 reagent kit. Genecards, Disgenet, and OMIM databases were used to identify potential disease-related targets. Molecular docking methods were performed to predict the binding interactions between compounds and core targets. Results We designed and synthesized compounds TB-1 to TB-16. Following the evaluation of their safety, TB-1, TB-2, TB-12, and TB-16 were selected for further investigation of their neuroprotective properties. The compound designed in this study exhibits a dose-dependent protective effect on glutamate-damaged PC12 cells. Further network pharmacology and molecular docking analyses indicate that TB-2 possesses a potential therapeutic effect against cerebral ischemia, and its possible targets were SRC, MAPK1 and KDR. Discussion The results indicated that TB-2 displayed a significant neuroprotective effect against Glu-induced injury in PC12 cells, suggesting potential therapeutic implications for cerebral ischemia.
Collapse
Affiliation(s)
- Lijuan Zhi
- Center Laboratory, Xi’an Mental Health Center, Xi’an, China
| | - Huan Li
- Center Laboratory, Xi’an Mental Health Center, Xi’an, China
| | - Baimei Shi
- College of Chemical Engineering, Xi’an University, Xi’an, China
| | - Tao Yu
- Center Laboratory, Xi’an Mental Health Center, Xi’an, China
| | - Xiaoni Jia
- Center Laboratory, Xi’an Mental Health Center, Xi’an, China
| | - Hui Zhang
- Center Laboratory, Xi’an Mental Health Center, Xi’an, China
| |
Collapse
|
22
|
Cortes Ballen AI, Amosu M, Ravinder S, Chan J, Derin E, Slika H, Tyler B. Metabolic Reprogramming in Glioblastoma Multiforme: A Review of Pathways and Therapeutic Targets. Cells 2024; 13:1574. [PMID: 39329757 PMCID: PMC11430559 DOI: 10.3390/cells13181574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive and highly malignant primary brain tumor characterized by rapid growth and a poor prognosis for patients. Despite advancements in treatment, the median survival time for GBM patients remains low. One of the crucial challenges in understanding and treating GBMs involves its remarkable cellular heterogeneity and adaptability. Central to the survival and proliferation of GBM cells is their ability to undergo metabolic reprogramming. Metabolic reprogramming is a process that allows cancer cells to alter their metabolism to meet the increased demands of rapid growth and to survive in the often oxygen- and nutrient-deficient tumor microenvironment. These changes in metabolism include the Warburg effect, alterations in several key metabolic pathways including glutamine metabolism, fatty acid synthesis, and the tricarboxylic acid (TCA) cycle, increased uptake and utilization of glutamine, and more. Despite the complexity and adaptability of GBM metabolism, a deeper understanding of its metabolic reprogramming offers hope for developing more effective therapeutic interventions against GBMs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; (A.I.C.B.); (M.A.); (S.R.); (J.C.); (E.D.); (H.S.)
| |
Collapse
|
23
|
Ghayee HK, Costa KA, Xu Y, Hatch HM, Rodriguez M, Straight SC, Bustamante M, Yu F, Smagulova F, Bowden JA, Tevosian SG. Polyamine Pathway Inhibitor DENSPM Suppresses Lipid Metabolism in Pheochromocytoma Cell Line. Int J Mol Sci 2024; 25:10029. [PMID: 39337514 PMCID: PMC11432427 DOI: 10.3390/ijms251810029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Pheochromocytomas (PCCs) are tumors arising from chromaffin cells in the adrenal medulla, and paragangliomas (PGLs) are tumors derived from extra-adrenal sympathetic or parasympathetic paraganglia; these tumors are collectively referred to as PPGL cancer. Treatment for PPGL primarily involves surgical removal of the tumor, and only limited options are available for treatment of the disease once it becomes metastatic. Human carriers of the heterozygous mutations in the succinate dehydrogenase subunit B (SDHB) gene are susceptible to the development of PPGL. A physiologically relevant PCC patient-derived cell line hPheo1 was developed, and SDHB_KD cells carrying a stable short hairpin knockdown of SDHB were derived from it. An untargeted metabolomic approach uncovered an overactive polyamine pathway in the SDHB_KD cells that was subsequently fully validated in a large set of human SDHB-mutant PPGL tumor samples. We previously reported that treatment with the polyamine metabolism inhibitor N1,N11-diethylnorspermine (DENSPM) drastically inhibited growth of these PCC-derived cells in culture as well as in xenograft mouse models. Here we explored the mechanisms underlying DENSPM action in hPheo1 and SDHB_KD cells. Specifically, by performing an RNAseq analysis, we have identified gene expression changes associated with DENSPM treatment that broadly interfere with all aspects of lipid metabolism, including fatty acid (FA) synthesis, desaturation, and import/uptake. Furthermore, by performing an untargeted lipidomic liquid chromatography-mass spectrometry (LC/MS)-based analysis we uncovered specific groups of lipids that are dramatically reduced as a result of DENSPM treatment. Specifically, the bulk of plasmanyl ether lipid species that have been recently reported as the major determinants of cancer cell fate are notably decreased. In summary, this work suggests an intersection between active polyamine and lipid pathways in PCC cells.
Collapse
Affiliation(s)
- Hans K. Ghayee
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL 32608, USA; (Y.X.); (M.B.)
| | - Kaylie A. Costa
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Yiling Xu
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL 32608, USA; (Y.X.); (M.B.)
| | - Heather M. Hatch
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Mateo Rodriguez
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Shelby C. Straight
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Marian Bustamante
- Department of Medicine, Division of Endocrinology, College of Medicine, University of Florida and Malcom Randall VA Medical Center, Gainesville, FL 32608, USA; (Y.X.); (M.B.)
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Fahong Yu
- The Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL 32610, USA;
| | - Fatima Smagulova
- Université de Rennes, EHESP, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), Campus Sante de Villejean—UMR_S 1085, F-35000 Rennes, France;
| | - John A. Bowden
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| | - Sergei G. Tevosian
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 03610, USA; (K.A.C.); (H.M.H.); (M.R.); (S.C.S.); (J.A.B.)
| |
Collapse
|
24
|
Wang X, Li Y, Hou X, Li J, Ma X. Lipid metabolism reprogramming in endometrial cancer: biological functions and therapeutic implications. Cell Commun Signal 2024; 22:436. [PMID: 39256811 PMCID: PMC11385155 DOI: 10.1186/s12964-024-01792-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Endometrial cancer is one of the major gynecological cancers, with increasing incidence and mortality in the past decades. Emerging preclinical and clinical data have indicated its close association with obesity and dyslipidemia. Metabolism reprogramming has been considered as the hallmark of cancer, to satisfy the extensive need of nutrients and energy for survival and growth. Particularly, lipid metabolism reprogramming has aroused the researchers' interest in the field of cancer, including tumorigenesis, invasiveness, metastasis, therapeutic resistance and immunity modulation, etc. But the roles of lipid metabolism reprogramming in endometrial cancer have not been fully understood. This review has summarized how lipid metabolism reprogramming induces oncogenesis and progression of endometrial cancer, including the biological functions of aberrant lipid metabolism pathway and altered transcription regulation of lipid metabolism pathway. Besides, we proposed novel therapeutic strategies of targeting lipid metabolism pathway and concentrated on its potential of sensitizing immunotherapy and hormonal therapy, to further optimize the existing treatment modalities of patients with advanced/metastatic endometrial cancer. Moreover, we expect that targeting lipid metabolism plus hormone therapy may block the endometrial malignant transformation and enrich the preventative approaches of endometrial cancer. CONCLUSION Lipid metabolism reprogramming plays an important role in tumor initiation and cancer progression of endometrial cancer. Targeting the core enzymes and transcriptional factors of lipid metabolism pathway alone or in combination with immunotherapy/hormone treatment is expected to decrease the tumor burden and provide promising treatment opportunity for patients with advanced/metastatic endometrial cancer.
Collapse
Affiliation(s)
- Xiangyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Yinuo Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Xin Hou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Jingfang Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, Hubei Province, 430030, China
| | - Xiangyi Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, Hubei Province, 430030, China.
| |
Collapse
|
25
|
Zhu Z, Zhang Y, Wang L, Geng H, Li M, Chen S, Wang X, Chen P, Sun C, Zhang C. Spatial Metabolomics Profiling Reveals Curcumin Induces Metabolic Reprogramming in Three-Dimensional Tumor Spheroids. Metabolites 2024; 14:482. [PMID: 39330489 PMCID: PMC11433860 DOI: 10.3390/metabo14090482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Curcumin is widely recognized for its diverse antitumor properties, ranging from breast cancer to many other types of cancers. However, its role in the tumor microenvironment remains to be elucidated. In this study, we established a 3D tumor spheroids model that can simulate the growth environment of tumor cells and visualized the antitumor metabolic alteration caused by curcumin using mass spectrometry imaging technology. Our results showed that curcumin not only exerts a profound impact on the growth and proliferation of breast cancer cells but in situ multivariate statistical analysis also reveals the significant effect on the overall metabolic profile of tumor spheroids. Meanwhile, our visualization map characterized curcumin metabolic processes of reduction and glucuronidation in tumor spheroids. More importantly, abnormal metabolic pathways related to lipid metabolism and polyamine metabolism were also remodeled at the metabolite and gene levels after curcumin intervention. These insights deepen our comprehension of the regulatory mechanism of curcumin on the tumor metabolic network, furnishing powerful references for antitumor treatment.
Collapse
Affiliation(s)
- Zihan Zhu
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yaqi Zhang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Lei Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Haoyuan Geng
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Min Li
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Shiping Chen
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Xiao Wang
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Panpan Chen
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chenglong Sun
- Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Chao Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
26
|
Bai R, Cui J. Regulation of fatty acid synthase on tumor and progress in the development of related therapies. Chin Med J (Engl) 2024; 137:1894-1902. [PMID: 38273440 PMCID: PMC11332710 DOI: 10.1097/cm9.0000000000002880] [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: 05/19/2023] [Indexed: 01/27/2024] Open
Abstract
ABSTRACT Fatty acid synthase (FASN) is an essential molecule in lipid metabolic pathways, which are crucial for cancer-related studies. Recent studies have focused on a comprehensive understanding of the novel and important regulatory effects of FASN on malignant biological behavior and immune-cell infiltration, which are closely related to tumor occurrence and development, immune escape, and immune response. FASN-targeting antitumor treatment strategies are being developed. Therefore, in this review, we focused on the effects of FASN on tumor and immune-cell infiltration and reviewed the progress of related anti-tumor therapy development.
Collapse
Affiliation(s)
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
27
|
Cao LQ, Xie Y, Fleishman JS, Liu X, Chen ZS. Hepatocellular carcinoma and lipid metabolism: Novel targets and therapeutic strategies. Cancer Lett 2024; 597:217061. [PMID: 38876384 DOI: 10.1016/j.canlet.2024.217061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/10/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is an increasingly prevalent disease that is associated with high and continually rising mortality rates. Lipid metabolism holds a crucial role in the pathogenesis of HCC, in which abnormalities pertaining to the delicate balance of lipid synthesis, breakdown, and storage, predispose for the pathogenesis of the nonalcoholic fatty liver disease (NAFLD), a disease precursor to HCC. If caught early enough, HCC treatment may be curative. In later stages, treatment is only halting the inevitable outcome of death, boldly prompting for novel drug discovery to provide a fighting chance for this patient population. In this review, we begin by providing a summary of current local and systemic treatments against HCC. From such we discuss hepatic lipid metabolism and highlight novel targets that are ripe for anti-cancer drug discovery. Lastly, we provide a targeted summary of current known risk factors for HCC pathogenesis, providing key insights that will be essential for rationalizing future development of anti-HCC therapeutics.
Collapse
Affiliation(s)
- Lu-Qi Cao
- Institute for Biotechnology, St. John's University, New York, NY, 11439, USA; College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Yuhao Xie
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Xuan Liu
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518034, China.
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John's University, New York, NY, 11439, USA; College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA.
| |
Collapse
|
28
|
Li B, Mi J, Yuan Q. Fatty acid metabolism-related enzymes in colorectal cancer metastasis: from biological function to molecular mechanism. Cell Death Discov 2024; 10:350. [PMID: 39103344 DOI: 10.1038/s41420-024-02126-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024] Open
Abstract
Colorectal cancer (CRC) is a highly aggressive and life-threatening malignancy that metastasizes in ~50% of patients, posing significant challenges to patient survival and treatment. Fatty acid (FA) metabolism regulates proliferation, immune escape, metastasis, angiogenesis, and drug resistance in CRC. FA metabolism consists of three pathways: de novo synthesis, uptake, and FA oxidation (FAO). FA metabolism-related enzymes promote CRC metastasis by regulating reactive oxygen species (ROS), matrix metalloproteinases (MMPs), angiogenesis and epithelial-mesenchymal transformation (EMT). Mechanistically, the PI3K/AKT/mTOR pathway, wnt/β-catenin pathway, and non-coding RNA signaling pathway are regulated by crosstalk of enzymes related to FA metabolism. Given the important role of FA metabolism in CRC metastasis, targeting FA metabolism-related enzymes and their signaling pathways is a potential strategy to treat CRC metastasis.
Collapse
Affiliation(s)
- Biao Li
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Jing Mi
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Qi Yuan
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
29
|
Zhang J, He J, Chen W, Chen G, Wang L, Liu Y, Wang Z, Yang M, Huang G, Yang Y, Ma W, Li Y. Single-cell RNA-binding protein pattern-mediated molecular subtypes depict the hallmarks of the tumor microenvironment in bladder urothelial carcinoma. ONCOLOGIE 2024; 26:657-669. [DOI: 10.1515/oncologie-2024-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Abstract
Objectives
Bladder carcinoma (BC) is a common malignancy of the urinary tract. As a new hallmark of cancer for drug therapy, RNA-binding proteins (RBPs) are key regulatory factors in alternative splicing events. This work is to uncover the relationship between BC and RBP in order to find drug targets in BC.
Methods
In this work, data from single-cell RNA-seq GSE1355337, PRJNA662018, and the TCGA-Bladder urothelial carcinoma (BLCA) cohorts are integrated to identify their relationships. A scoring system is constructed according to RBPs gene expression and patients’ survival. A network is constructed to analyze the alternative splicing events and RBP genes.
Results
A scoring system identified 321 RBPs significantly associated with the prognosis of patients. Subsequent typing of these RBP genes in two single-cell datasets demonstrated that most of the RBP genes had variable copy numbers. Three RBP clusters were identified. Using RBP genes as a signature in BC epithelial cells allows for differentiation between different grades of BC samples. The novel RBP genes-based subtype system reflects BC clinical staging. Notably, CellChat analysis revealed that the RBP genes-associated cell subtypes of T cells had extensive interactions with epithelial cells. Further analysis showed that the ligand-receptor pair MIF-CXCR4 mediated the communication between RBP-associated subtypes of BC epithelial cells and T cells.
Conclusions
Taken together, RBP genes are associated with BC progress and offer new indicators for precision medicine in BC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Urology Surgery , Affiliated Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Jiejie He
- Department of Surgical Oncology , Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Wen Chen
- Wuhan Ruixing Biotechnology Co. Ltd. , Wuhan , Hubei Province , China
| | - Guojun Chen
- Department of Urology Surgery , Affiliated Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Liang Wang
- Department of Gastrointestinal Oncology , Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Yuchan Liu
- Department of Gynecology and Obstetrics , Jingmen Central Hospital , Jingmen , Hubei Province , China
| | - Zhanjin Wang
- Medical College of Qinghai University , Xining , Qinghai Province , China
| | - Ming Yang
- Department of Medical Records and Statistic, Affiliated Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Guoyi Huang
- Wuhan Ruixing Biotechnology Co. Ltd. , Wuhan , Hubei Province , China
| | - Yongli Yang
- Department of Gynecology , Affiliated Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Wei Ma
- Department of Surgery , Affiliated Hospital of Qinghai University , Xining , Qinghai Province , China
| | - Yan Li
- Department of Gynecologic Oncology , Affiliated Hospital of Qinghai University and Affiliated Cancer Hospital of Qinghai University , Xining , Qinghai Province , China
| |
Collapse
|
30
|
Li H, Zhou T, Zhang Q, Yao Y, Hua T, Zhang J, Wang H. Characterization and validation of fatty acid metabolism-related genes predicting prognosis, immune infiltration, and drug sensitivity in endometrial cancer. Biotechnol Appl Biochem 2024; 71:909-928. [PMID: 38616327 DOI: 10.1002/bab.2586] [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: 10/30/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024]
Abstract
Endometrial cancer is considered to be the second most common tumor of the female reproductive system, and patients diagnosed with advanced endometrial cancer have a poor prognosis. The influence of fatty acid metabolism in the prognosis of patients with endometrial cancer remains unclear. We constructed a prognostic risk model using transcriptome sequencing data of endometrial cancer and clinical information of patients from The Cancer Genome Atlas (TCGA) database via least absolute shrinkage and selection operator regression analysis. The tumor immune microenvironment was analyzed using the CIBERSORT algorithm, followed by functional analysis and immunotherapy efficacy prediction by gene set variation analysis. The role of model genes in regulating endometrial cancer in vitro was verified by CCK-8, colony formation, wound healing, and transabdominal invasion assays, and verified in vivo by subcutaneous tumor transplantation in nude mice. A prognostic model containing 14 genes was constructed and validated in 3 cohorts and clinical samples. The results showed differences in the infiltration of immune cells between the high-risk and low-risk groups, and that the high-risk group may respond better to immunotherapy. Experiments in vitro confirmed that knockdown of epoxide hydrolase 2 (EPHX2) and acyl-CoA oxidase like (ACOXL) had an inhibitory effect on EC cells, as did overexpression of hematopoietic prostaglandin D synthase (HPGDS). The same results were obtained in experiments in vivo. Prognostic models related to fatty acid metabolism can be used for the risk assessment of endometrial cancer patients. Experiments in vitro and in vivo confirmed that the key genes HPGDS, EPHX2, and ACOXL in the prognostic model may affect the development of endometrial cancer.
Collapse
Affiliation(s)
- Haojia Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuwei Yao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Teng Hua
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Clinical Research Center of Cancer Immunotherapy, Wuhan, Hubei, China
| |
Collapse
|
31
|
Huang Y, Lin Y, Lan W, Huang C, Zhong C. GloEC: a hierarchical-aware global model for predicting enzyme function. Brief Bioinform 2024; 25:bbae365. [PMID: 39073830 DOI: 10.1093/bib/bbae365] [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: 04/13/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
The annotation of enzyme function is a fundamental challenge in industrial biotechnology and pathologies. Numerous computational methods have been proposed to predict enzyme function by annotating enzyme labels with Enzyme Commission number. However, the existing methods face difficulties in modelling the hierarchical structure of enzyme label in a global view. Moreover, they haven't gone entirely to leverage the mutual interactions between different levels of enzyme label. In this paper, we formulate the hierarchy of enzyme label as a directed enzyme graph and propose a hierarchy-GCN (Graph Convolutional Network) encoder to globally model enzyme label dependency on the enzyme graph. Based on the enzyme hierarchy encoder, we develop an end-to-end hierarchical-aware global model named GloEC to predict enzyme function. GloEC learns hierarchical-aware enzyme label embeddings via the hierarchy-GCN encoder and conducts deductive fusion of label-aware enzyme features to predict enzyme labels. Meanwhile, our hierarchy-GCN encoder is designed to bidirectionally compute to investigate the enzyme label correlation information in both bottom-up and top-down manners, which has not been explored in enzyme function prediction. Comparative experiments on three benchmark datasets show that GloEC achieves better predictive performance as compared to the existing methods. The case studies also demonstrate that GloEC is capable of effectively predicting the function of isoenzyme. GloEC is available at: https://github.com/hyr0771/GloEC.
Collapse
Affiliation(s)
- Yiran Huang
- School of Computer, Electronics and Information, Guangxi University, Nanning 530004, China
- Key Laboratory of Parallel, Distributed and Intelligent Computing in Guangxi Universities and Colleges, Guangxi University, Nanning 530004, China
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning 530004, China
| | - Yufu Lin
- School of Computer, Electronics and Information, Guangxi University, Nanning 530004, China
| | - Wei Lan
- School of Computer, Electronics and Information, Guangxi University, Nanning 530004, China
- Key Laboratory of Parallel, Distributed and Intelligent Computing in Guangxi Universities and Colleges, Guangxi University, Nanning 530004, China
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning 530004, China
| | - Cuiyu Huang
- College of Chemistry, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Nankai University, Tianjin 300071, China
| | - Cheng Zhong
- School of Computer, Electronics and Information, Guangxi University, Nanning 530004, China
- Key Laboratory of Parallel, Distributed and Intelligent Computing in Guangxi Universities and Colleges, Guangxi University, Nanning 530004, China
- Guangxi Key Laboratory of Multimedia Communications and Network Technology, Guangxi University, Nanning 530004, China
| |
Collapse
|
32
|
Cigliano A, Simile MM, Vidili G, Pes GM, Dore MP, Urigo F, Cossu E, Che L, Feo C, Steinmann SM, Ribback S, Pascale RM, Evert M, Chen X, Calvisi DF. Fatty Acid Synthase Promotes Hepatocellular Carcinoma Growth via S-Phase Kinase-Associated Protein 2/p27 KIP1 Regulation. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1160. [PMID: 39064589 PMCID: PMC11278665 DOI: 10.3390/medicina60071160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: Aberrant upregulation of fatty acid synthase (FASN), catalyzing de novo synthesis of fatty acids, occurs in various tumor types, including human hepatocellular carcinoma (HCC). Although FASN oncogenic activity seems to reside in its pro-lipogenic function, cumulating evidence suggests that FASN's tumor-supporting role might also be metabolic-independent. Materials and Methods: In the present study, we show that FASN inactivation by specific small interfering RNA (siRNA) promoted the downregulation of the S-phase kinase associated-protein kinase 2 (SKP2) and the consequent induction of p27KIP1 in HCC cell lines. Results: Expression levels of FASN and SKP2 directly correlated in human HCC specimens and predicted a dismal outcome. In addition, forced overexpression of SKP2 rendered HCC cells resistant to the treatment with the FASN inhibitor C75. Furthermore, FASN deletion was paralleled by SKP2 downregulation and p27KIP1 induction in the AKT-driven HCC preclinical mouse model. Moreover, forced overexpression of an SKP2 dominant negative form or a p27KIP1 non-phosphorylatable (p27KIP1-T187A) construct completely abolished AKT-dependent hepatocarcinogenesis in vitro and in vivo. Conclusions: In conclusion, the present data indicate that SKP2 is a critical downstream effector of FASN and AKT-dependent hepatocarcinogenesis in liver cancer, envisaging the possibility of effectively targeting FASN-positive liver tumors with SKP2 inhibitors or p27KIP1 activators.
Collapse
Affiliation(s)
- Antonio Cigliano
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Maria M. Simile
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Gianpaolo Vidili
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Giovanni M. Pes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Maria P. Dore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Francesco Urigo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Eleonora Cossu
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
| | - Claudio Feo
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Sara M. Steinmann
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, 17489 Greifswald, Germany;
| | - Rosa M. Pascale
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany; (E.C.); (S.M.S.); (M.E.)
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Fracisco, CA 94143, USA; (L.C.); (X.C.)
- Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Diego F. Calvisi
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy; (A.C.); (M.M.S.); (G.V.); (G.M.P.); (M.P.D.); (F.U.); (C.F.); (R.M.P.)
| |
Collapse
|
33
|
Peng R, Ma X, Jiang Z, Duan Y, Lv S, Jing W. Integrative analysis of Anoikis-related genes reveals that FASN is a novel prognostic biomarker and promotes the malignancy of bladder cancer via Wnt/β-catenin pathway. Heliyon 2024; 10:e34029. [PMID: 39071712 PMCID: PMC11283158 DOI: 10.1016/j.heliyon.2024.e34029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Bladder cancer (BC) exhibits diversity in clinical outcomes and is characterized by heterogeneity. Anoikis, a form of programmed cell death, plays a crucial role in facilitating tumor invasion and metastasis. This study comprehensively investigated the genetic landscape of BC progression, identifying 300 differentially expressed Anoikis-related genes (DE-ARGs) through in-depth analysis of the GSE13507 datasets. Functional enrichment analysis revealed associations with diverse diseases and biological processes. Employing machine learning algorithms, a logistic regression model based on nine marker genes demonstrated superior accuracy in distinguishing BC from normal samples. Validation in TCGA datasets highlighted the prognostic significance of LRP1, FASN, and SIRT6, suggesting their potential as cancer biomarkers. Particularly, FASN emerged as an independent prognostic indicator, regulating BC cell proliferation and metastasis through the Wnt/β-catenin pathway. The study provides crucial insights into altered genetic landscapes and potential therapeutic strategies for BC, emphasizing the significance of FASN in BC prognosis and progression.
Collapse
Affiliation(s)
- Ruoyu Peng
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Xiaohan Ma
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of Pregnancy, Zhengzhou, 450000, China
| | - Zhiyun Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Yu Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Shaogang Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| | - Wei Jing
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Key Laboratory of Laboratory Medicine of Henan, Zhengzhou, 450000, China
| |
Collapse
|
34
|
Deng H, Rao X, Zhang S, Chen L, Zong Y, Zhou R, Meng R, Dong X, Wu G, Li Q. Protein kinase CK2: An emerging regulator of cellular metabolism. Biofactors 2024; 50:624-633. [PMID: 38158592 DOI: 10.1002/biof.2032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/02/2023] [Indexed: 01/03/2024]
Abstract
The protein kinase casein kinase 2 (CK2) exerts its influence on the metabolism of three major cellular substances by phosphorylating essential protein molecules involved in various cellular metabolic pathways. These substances include hormones, especially insulin, rate-limiting enzymes, transcription factors of key genes, and cytokines. This regulatory role of CK2 is closely tied to important cellular processes such as cell proliferation and apoptosis. Additionally, tumor cells undergo metabolic reprogramming characterized by aerobic glycolysis, accelerated lipid β-oxidation, and abnormally active glutamine metabolism. In this context, CK2, which is overexpressed in various tumors, also plays a pivotal role. Hence, this review aims to summarize the regulatory mechanisms of CK2 in diverse metabolic pathways and tumor development, providing novel insights for the diagnosis, treatment, and prognosis of metabolism-related diseases and cancers.
Collapse
Affiliation(s)
- Huilin Deng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinrui Rao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Leichong Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianwen Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Miglionico R, Matera I, Ventola GM, Marchese G, Abruzzese V, Monné M, Ostuni A, Bisaccia F. Gene Expression Reprogramming by Citrate Supplementation Reduces HepG2 Cell Migration and Invasion. Int J Mol Sci 2024; 25:6509. [PMID: 38928215 PMCID: PMC11203947 DOI: 10.3390/ijms25126509] [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: 04/28/2024] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Citrate, which is obtained from oxaloacetate and acetyl-CoA by citrate synthase in mitochondria, plays a key role in both normal and cancer cell metabolism. In this work, we investigated the effect of 10 mM extracellular citrate supplementation on HepG2 cells. Gene expression reprogramming was evaluated by whole transcriptome analysis using gene set enrichment analysis (GSEA). The transcriptomic data were validated through analyzing changes in the mRNA levels of selected genes by qRT-PCR. Citrate-treated cells exhibited the statistically significant dysregulation of 3551 genes; 851 genes were upregulated and 822 genes were downregulated. GSEA identified 40 pathways affected by differentially expressed mRNAs. The most affected biological processes were related to lipid and RNA metabolism. Several genes of the cytochrome P450 family were upregulated in treated cells compared to controls, including the CYP3A5 gene, a tumor suppressor in hepatocellular carcinoma (HCC) that plays an important protective role in HCC metastasis. The citrate-induced dysregulation of cytochromes could both improve the effectiveness of chemotherapeutics used in combination and reduce the aggressiveness of tumors by diminishing cell migration and invasion.
Collapse
Affiliation(s)
- Rocchina Miglionico
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| | - Ilenia Matera
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| | | | - Giovanna Marchese
- Genomix4Life Srl, 84081 Baronissi, Italy; (G.M.V.); (G.M.)
- Genome Research Center for Health-CRGS, 84081 Baronissi, Italy
| | - Vittorio Abruzzese
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| | - Magnus Monné
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| | - Angela Ostuni
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| | - Faustino Bisaccia
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy; (R.M.); (I.M.); (V.A.); (M.M.)
| |
Collapse
|
36
|
Khan F, Elsori D, Verma M, Pandey S, Obaidur Rab S, Siddiqui S, Alabdallah NM, Saeed M, Pandey P. Unraveling the intricate relationship between lipid metabolism and oncogenic signaling pathways. Front Cell Dev Biol 2024; 12:1399065. [PMID: 38933330 PMCID: PMC11199418 DOI: 10.3389/fcell.2024.1399065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Lipids, the primary constituents of the cell membrane, play essential roles in nearly all cellular functions, such as cell-cell recognition, signaling transduction, and energy provision. Lipid metabolism is necessary for the maintenance of life since it regulates the balance between the processes of synthesis and breakdown. Increasing evidence suggests that cancer cells exhibit abnormal lipid metabolism, significantly affecting their malignant characteristics, including self-renewal, differentiation, invasion, metastasis, and drug sensitivity and resistance. Prominent oncogenic signaling pathways that modulate metabolic gene expression and elevate metabolic enzyme activity include phosphoinositide 3-kinase (PI3K)/AKT, MAPK, NF-kB, Wnt, Notch, and Hippo pathway. Conversely, when metabolic processes are not regulated, they can lead to malfunctions in cellular signal transduction pathways. This, in turn, enables uncontrolled cancer cell growth by providing the necessary energy, building blocks, and redox potentials. Therefore, targeting lipid metabolism-associated oncogenic signaling pathways could be an effective therapeutic approach to decrease cancer incidence and promote survival. This review sheds light on the interactions between lipid reprogramming and signaling pathways in cancer. Exploring lipid metabolism as a target could provide a promising approach for creating anticancer treatments by identifying metabolic inhibitors. Additionally, we have also provided an overview of the drugs targeting lipid metabolism in cancer in this review.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - Deena Elsori
- Faculty of Resilience, Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Shivam Pandey
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Samra Siddiqui
- Department of Health Service Management, College of Public Health and Health Informatics, University of Hail, Haʼil, Saudi Arabia
| | - Nadiyah M. Alabdallah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Haʼil, Saudi Arabia
| | - Pratibha Pandey
- Chitkara Centre for Research and Development, Chitkara University, Himachal Pradesh, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| |
Collapse
|
37
|
Johari NA, Sapi’i NA, Jiunn Hieng AL, Ab Latif N, Amran SI, Hasham R, Jemon K. In vitro and in silico evaluation of Andrographis paniculata ethanolic crude extracts on fatty acid synthase expression on breast cancer cells. Biomedicine (Taipei) 2024; 14:60-73. [PMID: 38939097 PMCID: PMC11204123 DOI: 10.37796/2211-8039.1444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 06/29/2024] Open
Abstract
Background Fatty acid synthase (FASN), a key rate-limiting enzyme in the fatty acid biosynthesis pathway has been identified to be overexpressed in breast cancer. This overexpression has been affiliated with poor prognosis and resistance to chemotherapeutics. Consequently, FASN has come into focus as an appealing potential target for breast cancer treatment. Available FASN inhibitors, however, are unstable and have been correlated with adverse side effects. Objective This present study aims to investigate the potential of Andrographis paniculata ethanolic crude extract (AP) as a potent FASN inhibitor in breast cancer cells. Materials & methods This study used MTT assay and flow cytometry analysis to measure cell viability and apoptosis following AP treatment (0-500 μg/mL). Furthermore, FASN protein expression was evaluated using immunocytochemistry whereas lipid droplet formation was quantified using Oil Red O staining. Literature-based identified AP phytochemicals were subjected to the prediction of molecular docking and ADMET properties. Results This study demonstrated that AP significantly reduced cell viability while inducing apoptosis in breast cancer cells. In addition, for the first time, exposure to AP was demonstrated to drastically reduce intracellular FASN protein expression and lipid droplet accumulation in EMT6 and MCF-7 breast cancer cells. Docking simulation analysis demonstrated AP phytochemicals may have exerted an inhibitory effect by targeting the FASN Thioesterase (TE) domain similarly to the known FASN inhibitor, Orlistat. Moreover, all AP phytochemicals also possessed drug-likeness properties which are in accordance with Lipinski's rule of five. Conclusions These results highlight the potential of A. paniculata ethanolic crude extract as a FASN inhibitor and hence might have the potential to be further developed as a potent chemotherapeutic drug for breast cancer treatment.
Collapse
Affiliation(s)
- Nur Amanina Johari
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Nur Anisa Sapi’i
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Alvin Lu Jiunn Hieng
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Nurriza Ab Latif
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Syazwani Itri Amran
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Rosnani Hasham
- Institute of Bioproduct Development, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
- Department of Bioprocess and Polymer Engineering, School of Chemical and Energy Engineering, Faculty of Engineering, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| | - Khairunadwa Jemon
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, 81310 Skudai, Johor,
Malaysia
| |
Collapse
|
38
|
Terry AR, Hay N. Emerging targets in lipid metabolism for cancer therapy. Trends Pharmacol Sci 2024; 45:537-551. [PMID: 38762377 PMCID: PMC11162322 DOI: 10.1016/j.tips.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/31/2024] [Accepted: 04/17/2024] [Indexed: 05/20/2024]
Abstract
Cancer cells perturb lipid metabolic pathways for a variety of pro-tumorigenic functions, and deregulated cellular metabolism is a hallmark of cancer cells. Although alterations in lipid metabolism in cancer cells have been appreciated for over 20 years, there are no FDA-approved cancer treatments that target lipid-related pathways. Recent advances pertaining to cancer cell fatty acid synthesis (FAS), desaturation, and uptake, microenvironmental and dietary lipids, and lipid metabolism of tumor-infiltrating immune cells have illuminated promising clinical applications for targeting lipid metabolism. This review highlights emerging pathways and targets for tumor lipid metabolism that may soon impact clinical treatment.
Collapse
Affiliation(s)
- Alexander R Terry
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, 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
|
39
|
Li S, Liu Z, Chen Q, Chen Y, Ji S. A novel fatty acid metabolism-related signature identifies MUC4 as a novel therapy target for esophageal squamous cell carcinoma. Sci Rep 2024; 14:12476. [PMID: 38816411 PMCID: PMC11139939 DOI: 10.1038/s41598-024-62917-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
Fatty acid metabolism has been identified as an emerging hallmark of cancer, which was closely associated with cancer prognosis. Whether fatty acid metabolism-related genes (FMGs) signature play a more crucial role in biological behavior of esophageal squamous cell carcinoma (ESCC) prognosis remains unknown. Thus, we aimed to identify a reliable FMGs signature for assisting treatment decisions and prognosis evaluation of ESCC. In the present study, we conducted consensus clustering analysis on 259 publicly available ESCC samples. The clinical information was downloaded from The Cancer Genome Atlas (TCGA, 80 ESCC samples) and Gene Expression Omnibus (GEO) database (GSE53625, 179 ESCC samples). A consensus clustering arithmetic was used to determine the FMGs molecular subtypes, and survival outcomes and immune features were evaluated among the different subtypes. Kaplan-Meier analysis and the receiver operating characteristic (ROC) was applied to evaluate the reliability of the risk model in training cohort, validation cohort and all cohorts. A nomogram to predict patients' 1-year, 3-year and 5-year survival rate was also studied. Finally, CCK-8 assay, wound healing assay, and transwell assay were implemented to evaluate the inherent mechanisms of FMGs for tumorigenesis in ESCC. Two subtypes were identified by consensus clustering, of which cluster 2 is preferentially associated with poor prognosis, lower immune cell infiltration. A fatty acid (FA) metabolism-related risk model containing eight genes (FZD10, TACSTD2, MUC4, PDLIM1, PRSS12, BAALC, DNAJA2 and ALOX12B) was established. High-risk group patients displayed worse survival, higher stromal, immune and ESTIMATE scores than in the low-risk group. Moreover, a nomogram revealed good predictive ability of clinical outcomes in ESCC patients. The results of qRT-PCR analysis revealed that the MUC4 and BAALC had high expression level, and FZD10, PDLIM1, TACSTD2, ALOX12B had low expression level in ESCC cells. In vitro, silencing MUC4 remarkably inhibited ESCC cell proliferation, invasion and migration. Our study fills the gap of FMGs signature in predicting the prognosis of ESCC patients. These findings revealed that cluster subtypes and risk model of FMGs had effects on survival prediction, and were expected to be the potential promising targets for ESCC.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Operating Room, Weifang Traditional Chinese Hospital, Weifang, China
| | - Zhengcao Liu
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, No.16 Baita Road, Suzhou, 215001, China
| | - Qingqing Chen
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, No.16 Baita Road, Suzhou, 215001, China
| | - Yuetong Chen
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, No.16 Baita Road, Suzhou, 215001, China
| | - Shengjun Ji
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, No.16 Baita Road, Suzhou, 215001, China.
| |
Collapse
|
40
|
Capuano A, D’Urso G, Gazzillo E, Lauro G, Chini MG, D’Auria MV, Ferraro MG, Iazzetti F, Irace C, Bifulco G, Casapullo A. Fatty Acid Synthase as Interacting Anticancer Target of the Terpenoid Myrianthic Acid Disclosed by MS-Based Proteomics Approaches. Int J Mol Sci 2024; 25:5918. [PMID: 38892106 PMCID: PMC11172900 DOI: 10.3390/ijms25115918] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
This research focuses on the target deconvolution of the natural compound myrianthic acid, a triterpenoid characterized by an ursane skeleton isolated from the roots of Myrianthus arboreus and from Oenothera maritima Nutt. (Onagraceae), using MS-based chemical proteomic techniques. Application of drug affinity responsive target stability (DARTS) and targeted-limited proteolysis coupled to mass spectrometry (t-LiP-MS) led to the identification of the enzyme fatty acid synthase (FAS) as an interesting macromolecular counterpart of myrianthic acid. This result, confirmed by comparison with the natural ursolic acid, was thoroughly investigated and validated in silico by molecular docking, which gave a precise picture of the interactions in the MA/FAS complex. Moreover, biological assays showcased the inhibitory activity of myrianthic acid against the FAS enzyme, most likely related to its antiproliferative activity towards tumor cells. Given the significance of FAS in specific pathologies, especially cancer, the myrianthic acid structural moieties could serve as a promising reference point to start the potential development of innovative approaches in therapy.
Collapse
Affiliation(s)
- Alessandra Capuano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gilda D’Urso
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Erica Gazzillo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
- PhD Program in Drug Discovery and Development, Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, C.da Fonte Lappone, 86090 Pesche, Italy
| | - Maria Valeria D’Auria
- Department of Pharmacy, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy;
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Federica Iazzetti
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Carlo Irace
- Biochem Lab, Department of Pharmacy, School of Medicine and Surgery, University of Naples “Federico II”, Via Domenico Montesano 49, 80131 Naples, Italy; (F.I.); (C.I.)
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| | - Agostino Casapullo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (A.C.); (E.G.); (G.L.); (G.B.); (A.C.)
| |
Collapse
|
41
|
Roy D, Dion E, Sepeda JA, Peng J, Lingam SR, Townsend K, Sas A, Sun W, Tedeschi A. α2δ1-mediated maladaptive sensory plasticity disrupts adipose tissue homeostasis following spinal cord injury. Cell Rep Med 2024; 5:101525. [PMID: 38663398 PMCID: PMC11148638 DOI: 10.1016/j.xcrm.2024.101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 01/13/2024] [Accepted: 04/02/2024] [Indexed: 05/23/2024]
Abstract
Spinal cord injury (SCI) increases the risk of cardiometabolic disorders, including hypertension, dyslipidemia, and insulin resistance. Not only does SCI lead to pathological expansion of adipose tissue, but it also leads to ectopic lipid accumulation in organs integral to glucose and insulin metabolism. The pathophysiological changes that underlie adipose tissue dysfunction after SCI are unknown. Here, we find that SCI exacerbates lipolysis in epididymal white adipose tissue (eWAT). Whereas expression of the α2δ1 subunit of voltage-gated calcium channels increases in calcitonin gene-related peptide-positive dorsal root ganglia neurons that project to eWAT, conditional deletion of the gene encoding α2δ1 in these neurons normalizes eWAT lipolysis after SCI. Furthermore, α2δ1 pharmacological blockade through systemic administration of gabapentin also normalizes eWAT lipolysis after SCI, preventing ectopic lipid accumulation in the liver. Thus, our study provides insight into molecular causes of maladaptive sensory processing in eWAT, facilitating the development of strategies to reduce metabolic and cardiovascular complications after SCI.
Collapse
Affiliation(s)
- Debasish Roy
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Elliot Dion
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jesse A Sepeda
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Juan Peng
- Center for Biostatistics and Bioinformatics, The Ohio State University, Columbus, OH 43210, USA
| | - Sai Rishik Lingam
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kristy Townsend
- Department of Neurological Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Sas
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Wenjing Sun
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Chronic Brain Injury Program, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
42
|
Zuo Q, Wu Y, Hu Y, Shao C, Liang Y, Chen L, Guo Q, Huang P, Chen Q. Targeting lipid reprogramming in the tumor microenvironment by traditional Chinese medicines as a potential cancer treatment. Heliyon 2024; 10:e30807. [PMID: 38765144 PMCID: PMC11101863 DOI: 10.1016/j.heliyon.2024.e30807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024] Open
Abstract
In the last ten years, there has been a notable rise in the study of metabolic abnormalities in cancer cells. However, compared to glucose or glutamine metabolism, less attention has been paid to the importance of lipid metabolism in tumorigenesis. Recent developments in lipidomics technologies have allowed for detailed analysis of lipid profiles within cancer cells and other cellular players present within the tumor microenvironment (TME). Traditional Chinese medicine (TCM) and its bioactive components have a long history of use in cancer treatments and are also being studied for their potential role in regulating metabolic reprogramming within TME. This review focuses on four core abnormalities altered by lipid reprogramming in cancer cells: de novo synthesis and exogenous uptake of fatty acids (FAs), upregulated fatty acid oxidation (FAO), cholesterol accumulation, which offer benefits for tumor growth and metastasis. The review also discusses how altered lipid metabolism impacts infiltrating immune cell function and phenotype as these interactions between cancer-stromal become more pronounced during tumor progression. Finally, recent literature is highlighted regarding how cancer cells can be metabolically reprogrammed by specific Chinese herbal components with potential therapeutic benefits related to lipid metabolic and signaling pathways.
Collapse
Affiliation(s)
- Qian Zuo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yingchao Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuyu Hu
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Cui Shao
- The First Affiliated Traditional Chinese Medicine Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqi Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liushan Chen
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| | - Qianqian Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ping Huang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qianjun Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
43
|
Cheng L, Li Z, Zheng Q, Yao Q. Correlation study of serum lipid levels and lipid metabolism-related genes in cervical cancer. Front Oncol 2024; 14:1384778. [PMID: 38779100 PMCID: PMC11109420 DOI: 10.3389/fonc.2024.1384778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Objective Lipid metabolism plays an important role in cancer. The aim of this study was to investigate the relationship between lipid metabolism and the development of cervical cancer, and to explore the prognostic significance of lipid metabolism-related genes in patients with cervical cancer. Methods Initially, we retrospectively collected data from 1589 cervical cancer patients treated at the Affiliated Hospital of Qingdao University, with 1589 healthy individuals from the physical examination center serving as the control group. The correlation between their serum lipid levels and cervical cancer was analyzed. Subsequently, leveraging public databases, we conducted comprehensive studies on lipid metabolism-related genes. Additionally, we analyzed RNA expression profiling and clinical information sourced from TCGA and GTEx databases. Finally, we established a prognostic model integrating 9 genes associated with lipid metabolism and generated a nomogram model using R. GO and KEGG were performed to explore the functions and pathways of lipid metabolism-related genes. Results Our findings revealed that patients with cervical cancer exhibited dyslipidemia, characterized by elevated levels of TC, TG, and LDL-C, alongside reduced HDL-C levels compared to controls (P<0.05). Interestingly, compared with early-stage patients, advanced patients had lower HDL-C level and higher LDL-C level. Regression analysis further highlighted high TC, TG, and LDL-C as significant risk factors for cervical cancer. Then a total of 188 lipid metabolism-related genes were identified and a prognostic signature based on 9 genes was established and validated. The results of the GO and KEGG functional analysis indicated that the lipid metabolism-related genes are primarily concentrated on pathways associated with fatty acid metabolism. Conclusion Our study underscores the varying degrees of dyslipidemia observed in patients with cervical cancer, emphasizing the relevance of serum lipids in disease development. Our prognostic riskScore model predicted the overall survival time of patients based on 9 genes associated with lipid metabolism. These 9 genes may be tumor biomarkers and new targets for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zhuo Li
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qingmei Zheng
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qin Yao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
44
|
Strachowska M, Robaszkiewicz A. Characteristics of anticancer activity of CBP/p300 inhibitors - Features of their classes, intracellular targets and future perspectives of their application in cancer treatment. Pharmacol Ther 2024; 257:108636. [PMID: 38521246 DOI: 10.1016/j.pharmthera.2024.108636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024]
Abstract
Due to the contribution of highly homologous acetyltransferases CBP and p300 to transcription elevation of oncogenes and other cancer promoting factors, these enzymes emerge as possible epigenetic targets of anticancer therapy. Extensive efforts in search for small molecule inhibitors led to development of compounds targeting histone acetyltransferase catalytic domain or chromatin-interacting bromodomain of CBP/p300, as well as dual BET and CBP/p300 inhibitors. The promising anticancer efficacy in in vitro and mice models led CCS1477 and NEO2734 to clinical trials. However, none of the described inhibitors is perfectly specific to CBP/p300 since they share similarity of a key functional domains with other enzymes, which are critically associated with cancer progression and their antagonists demonstrate remarkable clinical efficacy in cancer therapy. Therefore, we revise the possible and clinically relevant off-targets of CBP/p300 inhibitors that can be blocked simultaneously with CBP/p300 thereby improving the anticancer potential of CBP/p300 inhibitors and pharmacokinetic predicting data such as absorption, distribution, metabolism, excretion (ADME) and toxicity.
Collapse
Affiliation(s)
- Magdalena Strachowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland; University of Lodz, Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, Banacha 12 /16, 90-237 Lodz, Poland.
| | - Agnieszka Robaszkiewicz
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biophysics, Pomorska 141/143, 90-236 Lodz, Poland; Johns Hopkins University School of Medicine, Institute of Fundamental and Basic Research, 600 5(th) Street South, Saint Petersburg FL33701, United States of America.
| |
Collapse
|
45
|
Premaratne A, Basu S, Bagchi A, Zhou T, Feng Q, Lin CY. Liver X Receptor Ligand GAC0001E5 Downregulates Antioxidant Capacity and ERBB2/HER2 Expression in HER2-Positive Breast Cancer Cells. Cancers (Basel) 2024; 16:1651. [PMID: 38730603 PMCID: PMC11083021 DOI: 10.3390/cancers16091651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The HER2-positive subtype accounts for approximately one-fifth of all breast cancers. Insensitivity and development of acquired resistance to targeted therapies in some patients contribute to their poor prognosis. HER2 overexpression is associated with metabolic reprogramming, facilitating cancer cell growth and survival. Novel liver X receptor (LXR) ligand GAC0001E5 (1E5) has been shown to inhibit cancer cell proliferation by disrupting glutaminolysis and inducing oxidative stress. In this study, HER2-positive breast cancer cells were treated with 1E5 to determine their potential inhibitory effects and mechanisms of action in HER2-positive breast cancers. Similar to previous observations in other cancer types, 1E5 treatments inhibited LXR activity, expression, and cancer cell proliferation. Expression of fatty acid synthesis genes, including fatty acid synthase (FASN), was downregulated following 1E5 treatment, and results from co-treatment experiments with an FASN inhibitor suggest that the same pathway is targeted by 1E5. Treatments with 1E5 disrupted glutaminolysis and resulted in increased oxidative stress. Strikingly, HER2 transcript and protein levels were both significantly downregulated by 1E5. Taken together, these findings indicate the therapeutic potential of targeting HER2 overexpression and associated metabolic reprogramming via the modulation of LXR in HER2-positive breast cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Chin-Yo Lin
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77004, USA
| |
Collapse
|
46
|
Zou J, Dai Y, Xu G, Kai Y, Lan L, Zhang J, Wang Y. Identification of two distinct head and neck squamous cell carcinoma subtypes based on fatty acid metabolism-related signatures: Implications for immunotherapy and chemotherapy. Medicine (Baltimore) 2024; 103:e37824. [PMID: 38640298 PMCID: PMC11029997 DOI: 10.1097/md.0000000000037824] [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: 07/19/2023] [Revised: 03/06/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
The dysregulation of lipid metabolism is a critical factor in the initiation and progression of tumors. In this investigation, we aim to characterize the molecular subtypes of head and neck squamous cell carcinoma (HNSCC) based on their association with fatty acid metabolism and develop a prognostic risk model. The transcriptomic and clinical data about HNSCC were obtained from public databases. Clustering analysis was conducted on fatty acid metabolism genes (FAMG) associated with prognosis, utilizing the non-negative matrix factorization algorithm. The immune infiltration, response to immune therapy, and drug sensitivity between molecular subtypes were evaluated. Differential expression genes were identified between subtypes, and a prognostic model was constructed using Cox regression analyses. A nomogram for HNSCC was constructed and evaluated. Thirty FAMGs have been found to exhibit differential expression in HNSCC, out of which three are associated with HNSCC prognosis. By performing clustering analysis on these 3 genes, 2 distinct molecular subtypes of HNSCC were identified that exhibit significant heterogeneity in prognosis, immune landscape, and treatment response. Using a set of 7778 genes that displayed differential expression between the 2 molecular subtypes, a prognostic risk model for HNSCC was constructed comprising 11 genes. This model has the ability to stratify HNSCC patients into high-risk and low-risk groups, which exhibit significant differences in prognosis, immune infiltration, and immune therapy response. Moreover, our data suggest that this risk model is negatively correlated with B cells and most T cells, but positively correlated with macrophages, mast cells, and dendritic cells. Ultimately, we constructed a nomogram incorporating both the risk signature and radiotherapy, which has demonstrated exceptional performance in predicting prognosis for HNSCC patients. A molecular classification system and prognostic risk models were developed for HNSCC based on FAMGs. This study revealed the potential involvement of FAMGs in modulating tumor immune microenvironment and response to treatment.
Collapse
Affiliation(s)
- Jianjun Zou
- Department of Otolaryngology, Hangzhou Red Cross Hospital (Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine), Hangzhou, China
| | - Yanbi Dai
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| | - Guangbo Xu
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| | - Yilong Kai
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| | - Lingfeng Lan
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| | - Junkun Zhang
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| | - Yufeng Wang
- Department of Otolaryngology, The First People’s Hospital of Yuhang District (The First Affiliated Hospital, Zhejiang University School of Medicine, Liangzhu Branch), Hangzhou, China
| |
Collapse
|
47
|
Polverino F, Mora A. Alveolar Epithelial Cell Dysfunction in Idiopathic Pulmonary Fibrosis Linked to Lipid Alterations: Therapeutic Implications. Am J Respir Cell Mol Biol 2024; 70:233-234. [PMID: 38271680 PMCID: PMC11478126 DOI: 10.1165/rcmb.2023-0432ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Affiliation(s)
| | - Ana Mora
- Division of Pulmonary, Critical Care, and Sleep Medicine Ohio State University Columbus, Ohio
| |
Collapse
|
48
|
Köberlin MS, Fan Y, Liu C, Chung M, Pinto AFM, Jackson PK, Saghatelian A, Meyer T. A fast-acting lipid checkpoint in G1 prevents mitotic defects. Nat Commun 2024; 15:2441. [PMID: 38499565 PMCID: PMC10948896 DOI: 10.1038/s41467-024-46696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Lipid synthesis increases during the cell cycle to ensure sufficient membrane mass, but how insufficient synthesis restricts cell-cycle entry is not understood. Here, we identify a lipid checkpoint in G1 phase of the mammalian cell cycle by using live single-cell imaging, lipidome, and transcriptome analysis of a non-transformed cell. We show that synthesis of fatty acids in G1 not only increases lipid mass but extensively shifts the lipid composition to unsaturated phospholipids and neutral lipids. Strikingly, acute lowering of lipid synthesis rapidly activates the PERK/ATF4 endoplasmic reticulum (ER) stress pathway that blocks cell-cycle entry by increasing p21 levels, decreasing Cyclin D levels, and suppressing Retinoblastoma protein phosphorylation. Together, our study identifies a rapid anticipatory ER lipid checkpoint in G1 that prevents cells from starting the cell cycle as long as lipid synthesis is low, thereby preventing mitotic defects, which are triggered by low lipid synthesis much later in mitosis.
Collapse
Affiliation(s)
- Marielle S Köberlin
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Yilin Fan
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Chad Liu
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94111, USA
| | - Mingyu Chung
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Antonio F M Pinto
- Clayton Foundation Laboratories for Peptide Biology and Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology and Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
49
|
Jiang T, Qi J, Xue Z, Liu B, Liu J, Hu Q, Li Y, Ren J, Song H, Xu Y, Xu T, Fan R, Song J. The m 6A modification mediated-lncRNA POU6F2-AS1 reprograms fatty acid metabolism and facilitates the growth of colorectal cancer via upregulation of FASN. Mol Cancer 2024; 23:55. [PMID: 38491348 PMCID: PMC10943897 DOI: 10.1186/s12943-024-01962-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/19/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) have emerged as key players in tumorigenesis and tumour progression. However, the biological functions and potential mechanisms of lncRNAs in colorectal cancer (CRC) are unclear. METHODS The novel lncRNA POU6F2-AS1 was identified through bioinformatics analysis, and its expression in CRC patients was verified via qRT-PCR and FISH. In vitro and in vivo experiments, such as BODIPY staining, Oil Red O staining, triglyceride (TAG) assays, and liquid chromatography mass spectrometry (LC-MS) were subsequently performed with CRC specimens and cells to determine the clinical significance, and functional roles of POU6F2-AS1. Biotinylated RNA pull-down, RIP, Me-RIP, ChIP, and patient-derived organoid (PDO) culture assays were performed to confirm the underlying mechanism of POU6F2-AS1. RESULTS The lncRNA POU6F2-AS1 is markedly upregulated in CRC and associated with adverse clinicopathological features and poor overall survival in CRC patients. Functionally, POU6F2-AS1 promotes the growth and lipogenesis of CRC cells both in vitro and in vivo. Mechanistically, METTL3-induced m6A modification is involved in the upregulation of POU6F2-AS1. Furthermore, upregulated POU6F2-AS1 could tether YBX1 to the FASN promoter to induce transcriptional activation, thus facilitating the growth and lipogenesis of CRC cells. CONCLUSIONS Our data revealed that the upregulation of POU6F2-AS1 plays a critical role in CRC fatty acid metabolism and might provide a novel promising biomarker and therapeutic target for CRC.
Collapse
Affiliation(s)
- Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Junwen Qi
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Zhenyu Xue
- Department of Radiation Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Jianquan Liu
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Qihang Hu
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yuqiu Li
- Affiliated First Clinical College, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
- Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Jing Ren
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Hu Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Teng Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ruizhi Fan
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
50
|
Sarkar S, Roy D, Chatterjee B, Ghosh R. Clinical advances in analytical profiling of signature lipids: implications for severe non-communicable and neurodegenerative diseases. Metabolomics 2024; 20:37. [PMID: 38459207 DOI: 10.1007/s11306-024-02100-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/06/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Lipids play key roles in numerous biological processes, including energy storage, cell membrane structure, signaling, immune responses, and homeostasis, making lipidomics a vital branch of metabolomics that analyzes and characterizes a wide range of lipid classes. Addressing the complex etiology, age-related risk, progression, inflammation, and research overlap in conditions like Alzheimer's Disease, Parkinson's Disease, Cardiovascular Diseases, and Cancer poses significant challenges in the quest for effective therapeutic targets, improved diagnostic markers, and advanced treatments. Mass spectrometry is an indispensable tool in clinical lipidomics, delivering quantitative and structural lipid data, and its integration with technologies like Liquid Chromatography (LC), Magnetic Resonance Imaging (MRI), and few emerging Matrix-Assisted Laser Desorption Ionization- Imaging Mass Spectrometry (MALDI-IMS) along with its incorporation into Tissue Microarray (TMA) represents current advances. These innovations enhance lipidomics assessment, bolster accuracy, and offer insights into lipid subcellular localization, dynamics, and functional roles in disease contexts. AIM OF THE REVIEW The review article summarizes recent advancements in lipidomic methodologies from 2019 to 2023 for diagnosing major neurodegenerative diseases, Alzheimer's and Parkinson's, serious non-communicable cardiovascular diseases and cancer, emphasizing the role of lipid level variations, and highlighting the potential of lipidomics data integration with genomics and proteomics to improve disease understanding and innovative prognostic, diagnostic and therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW Clinical lipidomic studies are a promising approach to track and analyze lipid profiles, revealing their crucial roles in various diseases. This lipid-focused research provides insights into disease mechanisms, biomarker identification, and potential therapeutic targets, advancing our understanding and management of conditions such as Alzheimer's Disease, Parkinson's Disease, Cardiovascular Diseases, and specific cancers.
Collapse
Affiliation(s)
- Sutanu Sarkar
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Deotima Roy
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Bhaskar Chatterjee
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India
| | - Rajgourab Ghosh
- Amity Institute of Biotechnology (AIBNK), Amity University, Rajarhat, Newtown Action Area 2, Kolkata, 700135, West Bengal, India.
| |
Collapse
|