1
|
Yi J, Wang H, Deng Q, Huang C, Zhang L, Sun M, Ren J, Qu X. A bacteria-based bioorthogonal platform disrupts the flexible lipid homeostasis for potent metabolic therapy. Chem Sci 2025; 16:6014-6022. [PMID: 40070470 PMCID: PMC11891781 DOI: 10.1039/d4sc06481j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer cells exhibit altered metabolism and energetics, prominently reprogramming lipid metabolism to support tumor growth and progression, making it a promising target for cancer therapy. However, traditional genetic and pharmaceutical approaches for disrupting lipid metabolism face challenges due to the adaptability of tumor metabolism and potential side effects on normal tissues. Here, we present a bacteria-based bioorthogonal platform combining transition metal catalysts and Lactobacillus to disrupt the flexible lipid homeostasis in tumors. This platform activates glutamine transporter inhibitors in situ, targeting lipid synthesis in hypoxic tumor environments, while Lactobacillus inhibits lipid accumulation. By disrupting lipid metabolism and glutamine utilization, the present study proposes a safe and potent strategy for cancer therapy, with potential applications for other metabolic diseases.
Collapse
Affiliation(s)
- Jiadai Yi
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Huan Wang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
| | - Qingqing Deng
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Congcong Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Lu Zhang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
| | - Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun Jilin 130022 P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China Hefei Anhui 230026 P. R. China
| |
Collapse
|
2
|
Du P, Li Y, Han A, Wang M, Liu J, Piao Y, Chen L. β-lapachone suppresses carcinogenesis of cervical cancer via interaction with AKT1. Front Pharmacol 2025; 16:1509568. [PMID: 40051559 PMCID: PMC11882534 DOI: 10.3389/fphar.2025.1509568] [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: 10/11/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Cervical cancer is one of the most prevalent malignant tumors affecting women worldwide, and affected patients often face a poor prognosis due to its high drug resistance and recurrence rates. β-lapachone, a quinone compound originally extracted from natural plants, is an antitumor agent that specifically targets NQO1. Methods CC cells were treated with varying concentrations of β-lapachone to examine its effects on glucose metabolism, proliferation, metastasis, angiogenesis, and EMT in vitro. The targets and action pathways of β-lapachone were identified using network pharmacology and molecular docking, with KEGG pathway enrichment analysis. Its effects and toxicity were verified in vivo using a nude mouse xenograft model. Results β-lapachone significantly inhibited the proliferation and metastasis of cervical cancer cells by regulating glucose metabolism, reducing tumor angiogenesis, and suppressing epithelial-mesenchymal transition (EMT) in cells with high NQO1 expression. Furthermore, we identified the inactivation of the PI3K/AKT/mTOR pathway as the key mechanism underlying these effects. AKT1 was identified as a potential target of β-lapachone in modulating glucose metabolism and EMT in cervical cancer cells. Conclusion These findings suggest that β-lapachone inhibits the malignant progression of cervical cancer by targeting AKT1 to regulate glucose metabolism in NQO1-overexpressing cells, providing a theoretical basis for developing novel therapeutic strategies for cervical cancer.
Collapse
Affiliation(s)
- Pan Du
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Yue Li
- Changchun Center for Disease Control and Prevention, Changchun, China
| | - Anna Han
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Mengying Wang
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Jiajing Liu
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
| | - Yingshi Piao
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
- Cancer Research Center, Yanbian University, Yanji, China
| | - Liyan Chen
- Key Laboratory of Pathobiology (Yanbian University), State Ethnic Affairs Commission, Yanji, China
- Cancer Research Center, Yanbian University, Yanji, China
| |
Collapse
|
3
|
Rice A, Zourou AC, Goodell EP, Fu R, Pastor RW, Cotten ML. Investigating How Lysophosphatidylcholine and Lysophosphatidylethanolamine Enhance the Membrane Permeabilization Efficacy of Host Defense Peptide Piscidin 1. J Phys Chem B 2025; 129:210-227. [PMID: 39681296 PMCID: PMC11816835 DOI: 10.1021/acs.jpcb.4c05845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Lysophospholipids (LPLs) and host defense peptides (HDPs) are naturally occurring membrane-active agents that disrupt key membrane properties, including the hydrocarbon thickness, intrinsic curvature, and molecular packing. Although the membrane activity of these agents has been widely examined separately, their combined effects are largely unexplored. Here, we use experimental and computational tools to investigate how lysophosphatidylcholine (LPC) and lysophosphatidylethanolamine (LPE), an LPL of lower positive spontaneous curvature, influence the membrane activity of piscidin 1 (P1), an α-helical HDP from fish. Four membrane systems are probed: 75:25 C16:0-C18:1 PC (POPC)/C16:0-C18:1 phosphoglycerol (POPG), 50:25:25 POPC/POPG/16:0 LPC, 75:25 C16:0-C18:1 PE (POPE)/POPG, and 50:25:25 POPE/POPG/14:0 LPE. Dye leakage, circular dichroism, and NMR experiments demonstrate that while the presence of LPLs alone does not induce leakage-proficient defects, it boosts the permeabilization capability of P1, resulting in an efficacy order of POPC/POPG/16:0 LPC > POPE/POPG/14:0 LPE > POPC/POPG > POPE/POPG. This enhancement occurs without altering the membrane affinity and conformation of P1. Molecular dynamics simulations feature two types of asymmetric membranes to represent the imbalanced ("area stressed") and balanced ("area relaxed") distribution of lipids and peptides in the two leaflets. The simulations capture the membrane thinning effects of P1, LPC, and LPE, and the positive curvature strain imposed by both LPLs is reflected in the lateral pressure profiles. They also reveal a higher number of membrane defects for the P1/LPC than P1/LPE combination, congruent with the permeabilization experiments. Altogether, these results show that P1 and LPLs disrupt membranes in a concerted fashion, with LPC, the more disruptive LPL, boosting the permeabilization of P1 more than LPE. This mechanistic knowledge is relevant to understanding biological processes where multiple membrane-active agents such as HDPs and LPLs are involved.
Collapse
Affiliation(s)
- Amy Rice
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - Evan P. Goodell
- Department of Applied Science, William & Mary, Williamsburg, VA 23185
| | - Riqiang Fu
- National High Field Magnetic Laboratory, Tallahassee, FL, 32310
| | - Richard W. Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Myriam L. Cotten
- Department of Applied Science, William & Mary, Williamsburg, VA 23185
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR 97331
| |
Collapse
|
4
|
Zhao B, Yang J, Ran F, Shi Y, Yang L, Duan Y, Shi Z, Li X, Zhang J, Li Z, Wang J. CircBIRC6 affects prostate cancer progression by regulating miR-574-5p and DNAJB1. Cancer Biol Ther 2024; 25:2399363. [PMID: 39258752 PMCID: PMC11404571 DOI: 10.1080/15384047.2024.2399363] [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: 06/03/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is among the three main types of cancer. Although prostate-specific antigen (PSA) is routinely tested, it has disadvantages, such as poor prognostic ability. Therefore, finding more PCa markers and therapeutic targets remains a subject of study. CircRNAs have been found to have regulatory roles in various diseases, such as diabetes, Central Nervous System (CNS) neuropathy, etc. where their application in cancer is even more valuable. Therefore, this paper aims to search for differentially expressed circRNAs in PCa and find downstream targeting pathways related to autophagy. METHOD By detecting the expression of circRNA in the samples, hsa_circ_0119816 was finally identified as the research target. The properties of circRNA were verified by RNase R, actinomycin D, and fluorescence in situ hybridization (FISH). The downstream target miRNAs and target proteins were predicted by an online database, and the targeting relationship was verified using dual luciferase and RNA Immunoprecipitation. The effects of circRNAs and their downstream signalling pathways on prostate cancer cell proliferation, migration, EMT and autophagy were examined by CCK-8, Transwell, immunofluorescence and Western blotting. RESULTS CircBIRC6 is highly expressed in prostate cancer samples. Knockdown of its expression inhibits cell proliferation, invasion, EMT and autophagy and promotes apoptosis. CircBIRC6/miRNA-574-5p/DNAJB1 is a molecular axis that regulates prostate cancer cells.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Jinye Yang
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Fengming Ran
- Department of Pathology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Yuanlong Shi
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Libo Yang
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Yuanpeng Duan
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Zhiyu Shi
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Xin Li
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Jianpeng Zhang
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Zhiyao Li
- Department of Ultrasound Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
5
|
Calabrese C, Miserocchi G, De Vita A, Spadazzi C, Cocchi C, Vanni S, Gabellone S, Martinelli G, Ranallo N, Bongiovanni A, Liverani C. Lipids and adipocytes involvement in tumor progression with a focus on obesity and diet. Obes Rev 2024; 25:e13833. [PMID: 39289899 DOI: 10.1111/obr.13833] [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: 10/23/2023] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The adipose tissue is a complex organ that can play endocrine, metabolic, and immune regulatory roles in cancer. In particular, adipocytes provide metabolic substrates for cancer cell proliferation and produce signaling molecules that can stimulate cell adhesion, migration, invasion, angiogenesis, and inflammation. Cancer cells, in turn, can reprogram adipocytes towards a more inflammatory state, resulting in a vicious cycle that fuels tumor growth and evolution. These mechanisms are enhanced in obesity, which is associated with the risk of developing certain tumors. Diet, an exogenous source of lipids with pro- or anti-inflammatory functions, has also been connected to cancer risk. This review analyzes how adipocytes and lipids are involved in tumor development and progression, focusing on the relationship between obesity and cancer. In addition, we discuss how diets with varying lipid intakes can affect the disease outcomes. Finally, we introduce novel metabolism-targeted treatments and adipocyte-based therapies in oncology.
Collapse
Affiliation(s)
- Chiara Calabrese
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giacomo Miserocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandro De Vita
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Spadazzi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Cocchi
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Silvia Vanni
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sofia Gabellone
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicoletta Ranallo
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alberto Bongiovanni
- Clinical and Experimental Oncology, Immunotherapy, Rare Cancers and Biological Resource Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Liverani
- Preclinic and Osteoncology Unit, Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
6
|
Shamis SAK, Quinn J, Al‐Badran S, McKenzie M, Hatthakarnkul P, Lynch G, Lian G, Numprasit W, Romics L, Andersen D, Mallon E, McMillan DC, Edwards J. Elucidation of Dysregulated Pathways Associated With Hypoxia in Oestrogen Receptor-Negative Breast Cancer. Cancer Med 2024; 13:e70274. [PMID: 39660488 PMCID: PMC11632397 DOI: 10.1002/cam4.70274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/06/2024] [Accepted: 09/20/2024] [Indexed: 12/12/2024] Open
Abstract
PURPOSE Carbonic anhydrase IX (CAIX) is a well-established prognostic marker in breast cancer (BC). Nevertheless, this prognostic value is yet to be confirmed in BC subtypes. This study aims to investigate the prognostic effects of CAIX in oestrogen receptor (ER)-negative (ER-) BCs and to establish pathways related to cytoplasmic CAIX expression in ER- and lymph node-negative BCs. METHODS Immunohistochemistry was performed to identify the prognostic role of CAIX protein expression in ER- tissue microarrays (TMAs) (n = 191). CAIX-positive samples (n = 37) were transcriptionally profiled by TempO-Seq and analysed by STRING. Real-time quantitative PCR (RT-qPCR) analysis was used to validate differentially expressed genes. RESULTS Overexpression of cytoplasmic CAIX was an independent predictor of recurrence free survival, disease-free survival and overall survival in ER- cohort. RNA transcriptomic analysis identified 10 significant genes in ER- cohort and 3 genes in the node-negative group. The STRING database demonstrated a significant interaction between MUCL1 and GALNT6, which were linked with extracellular matrix organisation, degradation of the extracellular matrix and disease of glycosylation pathways. In the node-negative group, SPNS2 is mainly involved in the sphingolipid de novo biosynthesis pathway. A significant correlation between cytoplasmic SphK1 and cytoplasmic hypoxia-inducible factor-1α was observed. Among the 10 genes, 7 genes (SERHL2, GALNT6, MUCL1, MMP7, PITX2, CEACAM6 and SPNS2) were selected, and their expression was quantitatively assessed by RT-qPCR. The PCR data of these genes showed that SERHL2, GALNT6, MUCL1, PITX2, and SPNS2 mRNA levels were expressed in MDA-MB-231 BC cell lines at variable levels of hypoxic exposure. CONCLUSION Cytoplasmic CAIX was independently associated with poor prognosis in ER- BC. Gene expression profiles shed light on the pathways and genes associated with hypoxia in ER- BC. In node-negative patients, SPNS2 was of particular interest.
Collapse
Affiliation(s)
- Suad A. K. Shamis
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Jean Quinn
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Sara Al‐Badran
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Molly McKenzie
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Phimmada Hatthakarnkul
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Gerard Lynch
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Guang‐Yu Lian
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Warapan Numprasit
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Laszlo Romics
- NHS Greater Glasgow and Clyde (New Victoria Hospital)GlasgowUK
| | - Ditte Andersen
- BioClavis Ltd., Teaching & Learning CentreQueen Elizabeth University HospitalGlasgowUK
| | - Elizabeth Mallon
- Department of PathologyQueen Elizabeth University HospitalGlasgowUK
| | - Donald C. McMillan
- Academic Unit of Surgery, School of MedicineUniversity of Glasgow, Royal InfirmaryGlasgowUK
| | - Joanne Edwards
- Wolfson Wohl Cancer Research Centre, School of Cancer SciencesUniversity of GlasgowGlasgowUK
| |
Collapse
|
7
|
Lou Y, Jiang F, Guan J. The effect of lipidomes on the risk of endometrioid endometrial cancer: a Mendelian randomization study. Front Oncol 2024; 14:1436955. [PMID: 39493450 PMCID: PMC11527595 DOI: 10.3389/fonc.2024.1436955] [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: 05/22/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Objective This study aimed to explore the potential effects between various human plasma lipidomes and endometrioid endometrial cancer (EEC) by using Mendelian randomization (MR) methods. Methods This study designated a total of 179 human plasma lipidomes from the genome-wide association study (GWAS) database as the exposure variable. An EEC-related dataset from the GWAS (GCST006465) served as the outcome variable. MR analyses used the inverse variance-weighted method (IVW), MR-Egger, weighted median, simple mode, and weighted mode methods for regression calculations, accounting for possible biases induced by linkage disequilibrium and weak instrument variables. Any lipidomes failing to pass heterogeneity and horizontal pleiotropy tests were deemed to lack significant causal impact on the outcome. Results The results of IVW analysis disclosed that a variety of human plasma lipidomes (n = 15) exhibited a significant causal effect on EEC (p < 0.05). A subset of these lipidomes (n = 13) passed heterogeneity and horizontal pleiotropy tests, which demonstrated consistent and viable causal effects (p < 0.05) including glycerophospholipids, glycerolipids, and sterols. Specifically, phosphatidylcholine (odds ratio [OR]: 1.065-1.129, p < 0.05) exhibited a significant positive causal effect on the occurrence of EEC. Conversely, sterol ester (OR = 0.936, p = 0.007), diacylglycerol (OR = 0.914, p = 0.036), phosphatidylcholine (OR: 0.903-0.927, p < 0.05), phosphatidylethanolamine (OR = 0.907, p = 0.046) and triacylglycerol (OR: 0.880-0.924, p < 0.05) showed a notable negative causal association with EEC, suggesting their inhibitory effects on the EEC occurrence. Conclusions The study revealed that human plasma lipidomes have complex impacts on EEC through Mendelian randomization. This indicated that the diversity of structural changes in lipidomes could show different effects on subtypes and then affect EEC occurrence. Although these lipids had the potential to be promising biomarkers, they needed to be further clinically validated nevertheless.
Collapse
Affiliation(s)
- Yaochen Lou
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jun Guan
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
8
|
Jia W, Yuan J, Zhang J, Li S, Lin W, Cheng B. Bioactive sphingolipids as emerging targets for signal transduction in cancer development. Biochim Biophys Acta Rev Cancer 2024; 1879:189176. [PMID: 39233263 DOI: 10.1016/j.bbcan.2024.189176] [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: 06/06/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Sphingolipids, crucial components of cellular membranes, play a vital role in maintaining cellular structure and signaling integrity. Disruptions in sphingolipid metabolism are increasingly implicated in cancer development. Key bioactive sphingolipids, such as ceramides, sphingosine-1-phosphate (S1P), ceramide-1-phosphate (C1P), and glycosphingolipids, profoundly impact tumor biology. They influence the behavior of tumor cells, stromal cells, and immune cells, affecting tumor aggressiveness, angiogenesis, immune modulation, and extracellular matrix remodeling. Furthermore, abnormal expression of sphingolipids and their metabolizing enzymes modulates the secretion of tumor-derived extracellular vesicles (TDEs), which are key players in creating an immunosuppressive tumor microenvironment, remodeling the extracellular matrix, and facilitating oncogenic signaling within in situ tumors and distant pre-metastatic niches (PMNs). Understanding the role of sphingolipids in the biogenesis of tumor-derived extracellular vesicles (TDEs) and their bioactive contents can pave the way for new biomarkers in cancer diagnosis and prognosis, ultimately enhancing comprehensive tumor treatment strategies.
Collapse
Affiliation(s)
- Wentao Jia
- Department of General Practice, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China
| | - Jiaying Yuan
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Jinbo Zhang
- Department of Pharmacy, Tianjin Rehabilitation and Recuperation Center, Joint Logistics Support Force, Tianjin 300000, China
| | - Shu Li
- Department of Gastroenterology, Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201900, China
| | - Wanfu Lin
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| | - Binbin Cheng
- Oncology Department of Traditional Chinese Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China; Faculty of Traditional Chinese Medicine, Naval Medical University, Shanghai 200043, China.
| |
Collapse
|
9
|
Akdeniz YS, Özkan S. New markers in chronic obstructive pulmonary disease. Adv Clin Chem 2024; 123:1-63. [PMID: 39181619 DOI: 10.1016/bs.acc.2024.06.001] [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] [Indexed: 08/27/2024]
Abstract
Chronic obstructive pulmonary disease (COPD), a global healthcare and socioeconomic burden, is a multifaceted respiratory disorder that results in substantial decline in health status and life quality. Acute exacerbations of the disease contribute significantly to increased morbidity and mortality. Consequently, the identification of reliable and effective biomarkers for rapid diagnosis, prediction, and prognosis of exacerbations is imperative. In addition, biomarkers play a crucial role in monitoring responses to therapeutic interventions and exploring innovative treatment strategies. Although established markers such as CRP, fibrinogen and neutrophil count are routinely used, a universal marker is lacking. Fortunately, an increasing number of studies based on next generation analytics have explored potential biomarkers in COPD. Here we review those advances and the need for standardized validation studies in the appropriate clinical setting.
Collapse
Affiliation(s)
- Yonca Senem Akdeniz
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye.
| | - Seda Özkan
- Department of Emergency Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, İstanbul, Türkiye
| |
Collapse
|
10
|
Peng L, Du Q, Xiang L, Gu H, Luo H, Xu Z, He H, Xia B, Zhou Z, Wang Y, Chen Y. Adherence to the low-fat diet pattern reduces the risk of lung cancer in American adults aged 55 years and above: a prospective cohort study. J Nutr Health Aging 2024; 28:100240. [PMID: 38663125 DOI: 10.1016/j.jnha.2024.100240] [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: 11/15/2023] [Revised: 04/06/2024] [Accepted: 04/13/2024] [Indexed: 07/07/2024]
Abstract
OBJECTIVES There is little evidence on the association between low-fat dietary patterns and lung cancer risk among middle-aged and older adults. To fill this gap, we comprehensively investigated the association of adherence to a low-fat diet (LFD) and intake of different fat components including saturated, monounsaturated, and polyunsaturated fatty acids with incidence of lung cancer and its subtypes [non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC)] among adults aged 55 years and older. DESIGN A prospective cohort study with a mean follow-up time of 8.8 years. SETTING AND PARTICIPANTS This study used data from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. The study population included 98,459 PLCO participants age 55 and over at baseline who completed food frequency questionnaires providing detailed dietary information and had no history of cancer. METHODS Dietary intake was assessed using a validated food frequency questionnaire at baseline. A LFD score was calculated based on fat, protein, and carbohydrate intake as a percentage of total calories. Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between LFD score and intake of fat components (in quartiles) and incident lung cancer and its subtypes over follow-up. Restricted cubic spline analyses were conducted to examine possible nonlinear relationships. Subgroup analyses were performed to evaluate potential effect modifiers, and several sensitivity analyses were conducted to assess the stability of the findings. RESULTS During a follow-up of 869,807.9 person-years, 1,642 cases of lung cancer were observed, consisting of 1,408 (85.75%) cases of NSCLC and 234 (14.25%) cases of SCLC. The highest versus the lowest quartiles of the LFD score were found to be associated with a reduced risk of lung cancer (HR, 0.76; 95% CI, 0.66-0.89), NSCLC (HR, 0.79; 95% CI, 0.67-0.93), and SCLC (HR, 0.59; 95% CI, 0.38-0.92). The restricted cubic spline plots demonstrated a linear dose-response relationship between the LFD score and the risk of lung cancer as well as its subtypes. This risk reduction association for overall lung cancer was more pronounced in smokers (HR, 0.71; 95% CI, 0.60-0.84; P for interaction = 0.003). For fat components, high consumption of saturated fatty acids was associated with an increased lung cancer risk (HR, 1.35; 95% CI, 1.10-1.66), especially for SCLC (HR, 2.05; 95% CI, 1.20-3.53). No significant association was found between consumption of monounsaturated or polyunsaturated fatty acids and incident lung cancer and its subtypes. CONCLUSIONS Our findings suggest that adherence to LFD may reduce the lung cancer risk, particularly in smokers; while high saturated fatty acids consumption may increase lung cancer risk, especially for SCLC, among middle-aged and older adults in the US population.
Collapse
Affiliation(s)
- Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingqing Du
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoyun Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiquan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Boning Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Chen
- Health Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther 2024; 9:70. [PMID: 38531882 DOI: 10.1038/s41392-024-01777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular metabolism is an intricate network satisfying bioenergetic and biosynthesis requirements of cells. Relevant studies have been constantly making inroads in our understanding of pathophysiology, and inspiring development of therapeutics. As a crucial component of epigenetics at post-transcription level, RNA modification significantly determines RNA fates, further affecting various biological processes and cellular phenotypes. To be noted, immunometabolism defines the metabolic alterations occur on immune cells in different stages and immunological contexts. In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications, including N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), N7-methylguanosine (m7G), Pseudouridine (Ψ), adenosine-to-inosine (A-to-I) editing, which are relatively the most studied types. Then regulatory roles of these RNA modification on metabolism in diverse health and disease contexts are comprehensively described, categorized as glucose, lipid, amino acid, and mitochondrial metabolism. And we highlight the regulation of RNA modifications on immunometabolism, further influencing immune responses. Above all, we provide a thorough discussion about clinical implications of RNA modification in metabolism-targeted therapy and immunotherapy, progression of RNA modification-targeted agents, and its potential in RNA-targeted therapeutics. Eventually, we give legitimate perspectives for future researches in this field from methodological requirements, mechanistic insights, to therapeutic applications.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Clinical Medicine, Shandong University, Jinan, China
| | - Si-Qing Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Tian Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yun-Fei Fei
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Fei Wang
- Neurosurgical Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
12
|
Kondo M, Kumagai S, Nishikawa H. Metabolic advantages of regulatory T cells dictated by cancer cells. Int Immunol 2024; 36:75-86. [PMID: 37837615 DOI: 10.1093/intimm/dxad035] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 10/16/2023] Open
Abstract
Cancer cells employ glycolysis for their survival and growth (the "Warburg effect"). Consequently, surrounding cells including immune cells in the tumor microenvironment (TME) are exposed to hypoglycemic, hypoxic, and low pH circumstances. Since effector T cells depend on the glycolysis for their survival and functions, the metabolically harsh TME established by cancer cells is unfavorable, resulting in the impairment of effective antitumor immune responses. By contrast, immunosuppressive cells such as regulatory T (Treg) cells can infiltrate, proliferate, survive, and exert immunosuppressive functions in the metabolically harsh TME, indicating the different metabolic dependance between effector T cells and Treg cells. Indeed, some metabolites that are harmful for effector T cells can be utilized by Treg cells; lactic acid, a harmful metabolite for effector T cells, is available for Treg cell proliferation and functions. Deficiency of amino acids such as tryptophan and glutamine in the TME impairs effector T cell activation but increases Treg cell populations. Furthermore, hypoxia upregulates fatty acid oxidation via hypoxia-inducible factor 1α (HIF-1α) and promotes Treg cell migration. Adenosine is induced by the ectonucleotidases CD39 and CD73, which are strongly induced by HIF-1α, and reportedly accelerates Treg cell development by upregulating Foxp3 expression in T cells via A2AR-mediated signals. Therefore, this review focuses on the current views of the unique metabolism of Treg cells dictated by cancer cells. In addition, potential cancer combination therapies with immunotherapy and metabolic molecularly targeted reagents that modulate Treg cells in the TME are discussed to develop "immune metabolism-based precision medicine".
Collapse
Affiliation(s)
- Masaki Kondo
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Department of Neurosurgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Shogo Kumagai
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, Chiba 277-8577, Japan
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
13
|
Li C, Xiong Z, Han J, Nian W, Wang Z, Cai K, Gao J, Wang G, Tao K, Cai M. Identification of a lipid homeostasis-related gene signature for predicting prognosis, immunity, and chemotherapeutic effect in patients with gastric cancer. Sci Rep 2024; 14:2895. [PMID: 38316848 PMCID: PMC10844315 DOI: 10.1038/s41598-024-52647-7] [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: 10/16/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
Gastric cancer (GC) is one of the most common and deadliest cancers worldwide. Lipid homeostasis is essential for tumour development because lipid metabolism is one of the most important metabolic reprogramming pathways within tumours. Elucidating the mechanism of lipid homeostasis in GC might significantly improve treatment strategies and patient prognosis. GSE62254 was applied to construct a lipid homeostasis-related gene signature score (HGSscore) by multiple bioinformatic algorithms including weighted gene coexpression network analysis (WGCNA) and LASSO-Cox regression. A nomogram based on HGSscore and relevant clinical characteristics was constructed to predict the survival of patients with GC. TIMER and xCell were used to evaluate immune and stromal cell infiltration in the tumour microenvironment. Correlations between lipid homeostasis-related genes and chemotherapeutic efficacy were analysed in GSCAlite. RT‒qPCR and cell viability assays were applied to verify the findings in this study. HGSscore was constructed based on eighteen lipid homeostasis-related genes that were selected by WGCNA and LASSO-Cox regression. HGSscore was strongly associated with advanced TNM stage and showed satisfactory value in predicting GC prognosis in three independent cohorts. Furthermore, we found that HGSscore was associated with the tumour mutation burden (TMB) and immune/stromal cell infiltration, which are related to GC prognosis, indicating that lipid homeostasis impacts the formation of the tumour microenvironment (TME). With respect to the GSCAlite platform, PLOD2 and TGFB2 were shown to be positively related to chemotherapeutic resistance, while SLC10A7 was a favourable factor for chemotherapy efficacy. Cell viability assays showed that disrupted lipid homeostasis could attenuate GC cell viability. Moreover, RT‒qPCR revealed that lipid homeostasis could influence expression of specific genes. We identified a lipid homeostasis-related gene signature that correlated with survival, clinical characteristics, the TME, and chemotherapeutic efficacy in GC patients. This research provides a new perspective for improving prognosis and guiding individualized chemotherapy for patients with GC.
Collapse
Affiliation(s)
- Chao Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen Xiong
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinxin Han
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqi Nian
- Department of Oncology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailin Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Cai
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Louati K, Maalej A, Kolsi F, Kallel R, Gdoura Y, Borni M, Hakim LS, Zribi R, Choura S, Sayadi S, Chamkha M, Mnif B, Khemakhem Z, Boudawara TS, Boudawara MZ, Safta F. Differential Proteome Profiling Analysis under Pesticide Stress by the Use of a Nano-UHPLC-MS/MS Untargeted Proteomic-Based Approach on a 3D-Developed Neurospheroid Model: Identification of Protein Interactions, Prognostic Biomarkers, and Potential Therapeutic Targets in Human IDH Mutant High-Grade Gliomas. J Proteome Res 2023; 22:3534-3558. [PMID: 37651309 PMCID: PMC10629271 DOI: 10.1021/acs.jproteome.3c00395] [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: 07/03/2023] [Indexed: 09/02/2023]
Abstract
High-grade gliomas represent the most common group of infiltrative primary brain tumors in adults associated with high invasiveness, agressivity, and resistance to therapy, which highlights the need to develop potent drugs with novel mechanisms of action. The aim of this study is to reveal changes in proteome profiles under stressful conditions to identify prognostic biomarkers and altered apoptogenic pathways involved in the anticancer action of human isocitrate dehydrogenase (IDH) mutant high-grade gliomas. Our protocol consists first of a 3D in vitro developing neurospheroid model and then treatment by a pesticide mixture at relevant concentrations. Furthermore, we adopted an untargeted proteomic-based approach with high-resolution mass spectrometry for a comparative analysis of the differentially expressed proteins between treated and nontreated spheroids. Our analysis revealed that the majority of altered proteins were key members in glioma pathogenesis, implicated in the cellular metabolism, biological regulation, binding, and catalytic and structural activity and linked to many cascading regulatory pathways. Our finding revealed that grade-IV astrocytomas promote the downstream of the mitogen-activated-protein-kinases/extracellular-signal-regulated kinase (MAPK1/ERK2) pathway involving massive calcium influx. The gonadotrophin-releasing-hormone signaling enhances MAKP activity and may serve as a negative feedback compensating regulator. Thus, our study can pave the way for effective new therapeutic and diagnostic strategies to improve the overall survival.
Collapse
Affiliation(s)
- Kaouthar Louati
- Laboratory
of Pharmacology, Analytics and Galenic Drug Development- LR12ES09,
Faculty of Pharmacy, University of Monastir, Road Avicenne, 5000 Monastir, Tunisia
| | - Amina Maalej
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Fatma Kolsi
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Rim Kallel
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Yassine Gdoura
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Mahdi Borni
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Leila Sellami Hakim
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
| | - Rania Zribi
- Higher Institute
of Applied Studies to Humanities of Tunis (ISEAHT), University of Tunis, 11 Road of Jebel Lakdhar, 1005 Tunis, Tunisia
| | - Sirine Choura
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Sami Sayadi
- Biotechnology
Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, 2713 Doha, Qatar
| | - Mohamed Chamkha
- Laboratory
of Environmental Bioprocesses, Centre of
Biotechnology of Sfax, Road of Sidi-Mansour, P.O. Box 1177, 3018 Sfax, Tunisia
| | - Basma Mnif
- Department
of Bacteriology, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Zouheir Khemakhem
- Legal Medicine
Department, Habib Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Tahya Sellami Boudawara
- Laboratory
of Pathological Anatomy and Cytology, Habib
Bourguiba University Hospital, Road El Ain km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Mohamed Zaher Boudawara
- Department
of Neurosurgery, Habib Bourguiba University
Hospital, Road El Ain
km 1.5, Avenue of Ferdaous, 3089 Sfax, Tunisia
- Faculty of
Medicine, University of Sfax, Avenue of Majida Boulila, 3029 Sfax, Tunisia
| | - Fathi Safta
- Laboratory
of Pharmacology, Analytics and Galenic Drug Development- LR12ES09,
Faculty of Pharmacy, University of Monastir, Road Avicenne, 5000 Monastir, Tunisia
| |
Collapse
|
15
|
Jin HR, Wang J, Wang ZJ, Xi MJ, Xia BH, Deng K, Yang JL. Lipid metabolic reprogramming in tumor microenvironment: from mechanisms to therapeutics. J Hematol Oncol 2023; 16:103. [PMID: 37700339 PMCID: PMC10498649 DOI: 10.1186/s13045-023-01498-2] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023] Open
Abstract
Lipid metabolic reprogramming is an emerging hallmark of cancer. In order to sustain uncontrolled proliferation and survive in unfavorable environments that lack oxygen and nutrients, tumor cells undergo metabolic transformations to exploit various ways of acquiring lipid and increasing lipid oxidation. In addition, stromal cells and immune cells in the tumor microenvironment also undergo lipid metabolic reprogramming, which further affects tumor functional phenotypes and immune responses. Given that lipid metabolism plays a critical role in supporting cancer progression and remodeling the tumor microenvironment, targeting the lipid metabolism pathway could provide a novel approach to cancer treatment. This review seeks to: (1) clarify the overall landscape and mechanisms of lipid metabolic reprogramming in cancer, (2) summarize the lipid metabolic landscapes within stromal cells and immune cells in the tumor microenvironment, and clarify their roles in tumor progression, and (3) summarize potential therapeutic targets for lipid metabolism, and highlight the potential for combining such approaches with other anti-tumor therapies to provide new therapeutic opportunities for cancer patients.
Collapse
Affiliation(s)
- Hao-Ran Jin
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zi-Jing Wang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ming-Jia Xi
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Bi-Han Xia
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Deng
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| | - Jin-Lin Yang
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, No.37 Guoxue Road, Wuhou District, Chengdu, 610041, China.
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Torres W, Pérez JL, Díaz MP, D’Marco L, Checa-Ros A, Carrasquero R, Angarita L, Gómez Y, Chacín M, Ramírez P, Villasmil N, Durán-Agüero S, Cano C, Bermúdez V. The Role of Specialized Pro-Resolving Lipid Mediators in Inflammation-Induced Carcinogenesis. Int J Mol Sci 2023; 24:12623. [PMID: 37628804 PMCID: PMC10454572 DOI: 10.3390/ijms241612623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/10/2023] [Accepted: 04/15/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is a process involving cell mutation, increased proliferation, invasion, and metastasis. Over the years, this condition has represented one of the most concerning health problems worldwide due to its significant morbidity and mortality. At present, the incidence of cancer continues to grow exponentially. Thus, it is imperative to open new avenues in cancer research to understand the molecular changes driving DNA transformation, cell-to-cell interaction derangements, and immune system surveillance decay. In this regard, evidence supports the relationship between chronic inflammation and cancer. In light of this, a group of bioactive lipids derived from polyunsaturated fatty acids (PUFAs) may have a position as novel anti-inflammatory molecules known as the specialized pro-resolving mediators (SPMs), a group of pro-resolutive inflammation agents that could improve the anti-tumor immunity. These molecules have the potential role of chemopreventive and therapeutic agents for various cancer types, and their effects have been documented in the scientific literature. Thus, this review objective centers around understanding the effect of SPMs on carcinogenesis and their potential therapeutic effect.
Collapse
Affiliation(s)
- Wheeler Torres
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - José Luis Pérez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - María P. Díaz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Luis D’Marco
- Grupo de Investigación en Enfermedades Cardiorrenales y Metabólicas, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Valencia, Spain
| | - Ana Checa-Ros
- Grupo de Investigación en Enfermedades Cardiorrenales y Metabólicas, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Valencia, Spain
| | - Rubén Carrasquero
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Lissé Angarita
- Escuela de Nutrición y Dietética, Facultad de Medicina, Universidad Andres Bello, Concepción 4260000, Chile
| | - Yosselin Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Maricarmen Chacín
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080022, Colombia
| | - Paola Ramírez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Nelson Villasmil
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Samuel Durán-Agüero
- Facultad de Ciencias Para el Cuidado de la Salud, Universidad San Sebastián, Los Leones 8420524, Chile
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080022, Colombia
| |
Collapse
|
17
|
Lee-Rueckert M, Canyelles M, Tondo M, Rotllan N, Kovanen PT, Llorente-Cortes V, Escolà-Gil JC. Obesity-induced changes in cancer cells and their microenvironment: Mechanisms and therapeutic perspectives to manage dysregulated lipid metabolism. Semin Cancer Biol 2023; 93:36-51. [PMID: 37156344 DOI: 10.1016/j.semcancer.2023.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/05/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Obesity has been closely related to cancer progression, recurrence, metastasis, and treatment resistance. We aim to review recent progress in the knowledge on the obese macroenvironment and the generated adipose tumor microenvironment (TME) inducing lipid metabolic dysregulation and their influence on carcinogenic processes. Visceral white adipose tissue expansion during obesity exerts systemic or macroenvironmental effects on tumor initiation, growth, and invasion by promoting inflammation, hyperinsulinemia, growth-factor release, and dyslipidemia. The dynamic relationship between cancer and stromal cells of the obese adipose TME is critical for cancer cell survival and proliferation as well. Experimental evidence shows that secreted paracrine signals from cancer cells can induce lipolysis in cancer-associated adipocytes, causing them to release free fatty acids and acquire a fibroblast-like phenotype. Such adipocyte delipidation and phenotypic change is accompanied by an increased secretion of cytokines by cancer-associated adipocytes and tumor-associated macrophages in the TME. Mechanistically, the availability of adipose TME free fatty acids and tumorigenic cytokines concomitant with the activation of angiogenic processes creates an environment that favors a shift in the cancer cells toward an aggressive phenotype associated with increased invasiveness. We conclude that restoring the aberrant metabolic alterations in the host macroenvironment and in adipose TME of obese subjects would be a therapeutic option to prevent cancer development. Several dietary, lipid-based, and oral antidiabetic pharmacological therapies could potentially prevent tumorigenic processes associated with the dysregulated lipid metabolism closely linked to obesity.
Collapse
Affiliation(s)
| | - Marina Canyelles
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Mireia Tondo
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Noemi Rotllan
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | | | - Vicenta Llorente-Cortes
- Wihuri Research Institute, Helsinki, Finland; Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain; CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain.
| | - Joan Carles Escolà-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau, Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| |
Collapse
|
18
|
Zhang B, Zhang L, Qi P, Pang R, Wang Z, Liu X, Shi Q, Zhang Q. Potential role of LPAR5 gene in prognosis and immunity of thyroid papillary carcinoma and pan-cancer. Sci Rep 2023; 13:5850. [PMID: 37037831 PMCID: PMC10086052 DOI: 10.1038/s41598-023-32733-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
Papillary carcinomas account for the largest proportion of thyroid cancers, with papillary thyroid carcinoma (PTC) being prone to early lymph node metastasis. Some studies have confirmed that LPAR5 can promote the progression of PTC, but immune-related analyses of LPAR5 and PTC have not been widely discussed. This study aimed to determine the role of LPAR5 in PTC prognosis and immunity. We will further explore the role of LPAR5 in 33 different tumor types. Regarding PTC, we analyzed the effect of LPAR5 expression on overall survival (OS). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed. Immune-related analyses of immune checkpoints (ICPs) and immune cell infiltration were also performed. For pan-cancer, R packages were used to analyze prognosis, tumor mutational burden (TMB), microsatellite instability (MSI), and immune cell infiltration. Analysis of tumor microenvironment (TME) and ICPs was performed using Sangerbox ( http://vip.sangerbox.com/home.html ). The TISIDB database ( http://cis.hku.hk/TISIDB/index.php ) was used to identify immune and molecular subtypes. LPAR5 expression is associated with PTC prognosis and immunity as well as various human tumors. LPAR5 may be a potential biomarker for multiple malignancies and may provide a new target for cancer immunotherapy.
Collapse
Affiliation(s)
- Ben Zhang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Lixi Zhang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Peng Qi
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Renzhu Pang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Ziming Wang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Xuyao Liu
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Qi Shi
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China
| | - Qiang Zhang
- Thyroid Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, 130000, Jilin Province, China.
| |
Collapse
|
19
|
Zhao X, Amevor FK, Cui Z, Wan Y, Xue X, Peng C, Li Y. Steatosis in metabolic diseases: A focus on lipolysis and lipophagy. Biomed Pharmacother 2023; 160:114311. [PMID: 36764133 DOI: 10.1016/j.biopha.2023.114311] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Fatty acids (FAs), as part of lipids, are involved in cell membrane composition, cellular energy storage, and cell signaling. FAs can also be toxic when their concentrations inside and/or outside the cell exceed physiological levels, which is called "lipotoxicity", and steatosis is a form of lipotoxity. To facilitate the storage of large quantities of FAs in cells, they undergo a process called lipolysis or lipophagy. This review focuses on the effects of lipolytic enzymes including cytoplasmic "neutral" lipolysis, lysosomal "acid" lipolysis, and lipophagy. Moreover, the impact of related lipolytic enzymes on lipid metabolism homeostasis and energy conservation, as well as their role in lipid-related metabolic diseases. In addition, we describe how they affect lipid metabolism homeostasis and energy conservation in lipid-related metabolic diseases with a focus on hepatic steatosis and cancer and the pathogenesis and therapeutic targets of AMPK/SIRTs/FOXOs, PI3K/Akt, PPARs/PGC-1α, MAPK/ERK1/2, TLR4/NF-κB, AMPK/mTOR/TFEB, Wnt/β-catenin through immune inflammation, oxidative stress and autophagy-related pathways. As well as the current application of lipolytic enzyme inhibitors (especially Monoacylglycerol lipase (MGL) inhibitors) to provide new strategies for future exploration of metabolic programming in metabolic diseases.
Collapse
Affiliation(s)
- Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Ministry of Education, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
20
|
Yan J, Chen Y, Luo M, Hu X, Li H, Liu Q, Zou Z. Chronic stress in solid tumor development: from mechanisms to interventions. J Biomed Sci 2023; 30:8. [PMID: 36707854 PMCID: PMC9883141 DOI: 10.1186/s12929-023-00903-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
Chronic stress results in disturbances of body hormones through the neuroendocrine system. Cancer patients often experience recurrent anxiety and restlessness during disease progression and treatment, which aggravates disease progression and hinders treatment effects. Recent studies have shown that chronic stress-regulated neuroendocrine systems secret hormones to activate many signaling pathways related to tumor development in tumor cells. The activated neuroendocrine system acts not only on tumor cells but also modulates the survival and metabolic changes of surrounding non-cancerous cells. Current clinical evidences also suggest that chronic stress affects the outcome of cancer treatment. However, in clinic, there is lack of effective treatment for chronic stress in cancer patients. In this review, we discuss the main mechanisms by which chronic stress regulates the tumor microenvironment, including functional regulation of tumor cells by stress hormones (stem cell-like properties, metastasis, angiogenesis, DNA damage accumulation, and apoptotic resistance), metabolic reprogramming and immune escape, and peritumor neuromodulation. Based on the current clinical treatment framework for cancer and chronic stress, we also summarize pharmacological and non-pharmacological therapeutic approaches to provide some directions for cancer therapy.
Collapse
Affiliation(s)
- Jiajing Yan
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Yibing Chen
- grid.207374.50000 0001 2189 3846Department of Gynecology and Obstetrics, First Affiliated Hospital, Genetic and Prenatal Diagnosis Center, Zhengzhou University, Zhengzhou, 450001 China
| | - Minhua Luo
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Xinyu Hu
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| | - Hongsheng Li
- grid.410737.60000 0000 8653 1072Department of Breast Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095 China
| | - Quentin Liu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510631 China ,grid.411971.b0000 0000 9558 1426Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044 Liaoning China
| | - Zhengzhi Zou
- grid.263785.d0000 0004 0368 7397MOE Key Laboratory of Laser Life Science & Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631 China ,grid.263785.d0000 0004 0368 7397Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631 China
| |
Collapse
|
21
|
Huang W, Zhao Y, Xu Z, Wu X, Qiao M, Zhu Z, Zhao Z. The Regulatory Mechanism of miR-574-5p Expression in Cancer. Biomolecules 2022; 13:biom13010040. [PMID: 36671425 PMCID: PMC9855975 DOI: 10.3390/biom13010040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) are a group of small, single-stranded, non-coding RNAs approximately 22 nucleotides in length. The dysregulation of miRNAs has been widely investigated in various pathological processes, including tumorigenesis, providing a biomarker for cancer diagnosis and prognosis. As a member of the miRNA family, miR-574-5p is located on the human chromosome 4p14 and is highly correlated with a high incidence of human cancers. Functional pathways as well as underlying novel mechanisms upregulate or downregulate miR-574-5p, which plays an important regulatory role in tumorigenesis and progression. In this review, we systematically summarize the context-dependent implications of miR-574-5p and review differences in miR-574-5p expression in cancer. We also investigate the intricate functions exerted by miR-574-5p in diverse pathological processes and highlight regulatory pathways, networks, and other underlying novel mechanisms. The clinical applications of miR-574-5p as a diagnostic biomarker, prognostic biomarker, and therapeutic mechanism are also discussed in this paper. On this basis, we anticipate that miR-574-5p will be a promising and effective biomarker and therapeutic target.
Collapse
|
22
|
Martin-Perez M, Urdiroz-Urricelqui U, Bigas C, Benitah SA. The role of lipids in cancer progression and metastasis. Cell Metab 2022; 34:1675-1699. [PMID: 36261043 DOI: 10.1016/j.cmet.2022.09.023] [Citation(s) in RCA: 213] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Lipids have essential biological functions in the body (e.g., providing energy storage, acting as a signaling molecule, and being a structural component of membranes); however, an excess of lipids can promote tumorigenesis, colonization, and metastatic capacity of tumor cells. To metastasize, a tumor cell goes through different stages that require lipid-related metabolic and structural adaptations. These adaptations include altering the lipid membrane composition for invading other niches and overcoming cell death mechanisms and promoting lipid catabolism and anabolism for energy and oxidative stress protective purposes. Cancer cells also harness lipid metabolism to modulate the activity of stromal and immune cells to their advantage and to resist therapy and promote relapse. All this is especially worrying given the high fat intake in Western diets. Thus, metabolic interventions aiming to reduce lipid availability to cancer cells or to exacerbate their metabolic vulnerabilities provide promising therapeutic opportunities to prevent cancer progression and treat metastasis.
Collapse
Affiliation(s)
- Miguel Martin-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, University of Barcelona, 08028 Barcelona, Spain.
| | - Uxue Urdiroz-Urricelqui
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Claudia Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain.
| |
Collapse
|
23
|
Chen Y, Yuan H, Yu Q, Pang J, Sheng M, Tang W. Bioinformatics Analysis and Structure of Gastric Cancer Prognosis Model Based on Lipid Metabolism and Immune Microenvironment. Genes (Basel) 2022; 13:genes13091581. [PMID: 36140749 PMCID: PMC9498347 DOI: 10.3390/genes13091581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The reprogramming of lipid metabolism is a new trait of cancers. However, the role of lipid metabolism in the tumor immune microenvironment (TIME) and the prognosis of gastric cancer remains unclear. METHODS Consensus clustering was applied to identify novel subgroups. ESTIMATE, TIMER, and MCPcounter algorithms were used to determine the TIME of the subgroups. The underlying mechanisms were elucidated using functional analysis. The prognostic model was established using the LASSO algorithm and multivariate Cox regression analysis. RESULTS Three molecular subgroups with significantly different survival were identified. The subgroup with relatively low lipid metabolic expression had a lower immune score and immune cells. The differentially expressed genes (DEGs) were concentrated in immune biological processes and cell migration via GO and KEGG analyses. GSEA analysis showed that the subgroups were mainly enriched in arachidonic acid metabolism. Gastric cancer survival can be predicted using risk models based on lipid metabolism genes. CONCLUSIONS The TIME of gastric cancer patients is related to the expression of lipid metabolism genes and could be used to predict cancer prognosis accurately.
Collapse
|
24
|
Uranbileg B, Kurano M, Kano K, Sakai E, Arita J, Hasegawa K, Nishikawa T, Ishihara S, Yamashita H, Seto Y, Ikeda H, Aoki J, Yatomi Y. Sphingosine 1-phosphate lyase facilitates cancer progression through converting sphingolipids to glycerophospholipids. Clin Transl Med 2022; 12:e1056. [PMID: 36125914 PMCID: PMC9488530 DOI: 10.1002/ctm2.1056] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/23/2022] [Accepted: 08/29/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND In addition to potent agonist properties for sphingosine 1-phosphate (S1P) receptors, intracellularly, S1P is an intermediate in metabolic conversion pathway from sphingolipids to glycerolysophospholipids (glyceroLPLs). We hypothesized that this S1P metabolism and its products might possess some novel roles in the pathogenesis of cancer, where S1P lyase (SPL) is a key enzyme. METHODS The mRNA levels of sphingolipid-related and other cancer-related factors were measured in human hepatocellular carcinoma (HCC), colorectal cancer, and esophageal cancer patients' tumours and in their adjacent non-tumour tissues. Phospholipids (PL) and glyceroLPLs were measured by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In-vitro experiments were performed in Colon 26 cell line with modulation of the SPL and GPR55 expressions. Xenograft model was used for determination of the cancer progression and for pharmacological influence. RESULTS Besides high SPL levels in human HCC and colon cancer, SPL levels were specifically and positively linked with levels of glyceroLPLs, including lysophosphatidylinositol (LPI). Overexpression of SPL in Colon 26 cells resulted in elevated levels of LPI and lysophosphatidylglycerol (LPG), which are agonists of GPR55. SPL overexpression-enhanced cell proliferation was inhibited by GPR55 silencing. Conversely, inhibition of SPL led to the opposite outcome and reversed by adding LPI, LPG, and metabolites generated during S1P degradation, which is regulated by SPL. The xenograft model results suggested the contribution of SPL and glyceroLPLs to tumour progression depending on levels of SPL and GPR55. Moreover, the pharmacological inhibition of SPL prevented the progression of cancer. The underlying mechanisms for the SPL-mediated cancer progression are the activation of p38 and mitochondrial function through the LPI, LPG-GPR55 axis and the suppression of autophagy in a GPR55-independent manner. CONCLUSION A new metabolic pathway has been proposed here in HCC and colon cancer, SPL converts S1P to glyceroLPLs, mainly to LPI and LPG, and facilitates cancer development.
Collapse
Affiliation(s)
- Baasanjav Uranbileg
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kuniyuki Kano
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Eri Sakai
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Arita
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nishikawa
- Surgical Oncology and Vascular Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Soichiro Ishihara
- Surgical Oncology and Vascular Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Hiroharu Yamashita
- Gastrointestinal Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan.,Division of Digestive Surgery, Department of Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Yasuyuki Seto
- Gastrointestinal Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Ikeda
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junken Aoki
- Department of Health Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Liu JC, Zhang CL, Dong KY, Li MJ, Sun SG, Li CR. Advances in the research of plant-derived natural products against retinoblastoma. Int J Ophthalmol 2022; 15:1391-1400. [PMID: 36017045 DOI: 10.18240/ijo.2022.08.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/24/2021] [Indexed: 11/23/2022] Open
Abstract
Retinoblastoma (RB) is a highly aggressive ocular tumor, and due to socioeconomic and medical constraints, many children receive treatment only in the metaphase and advanced clinical stages, resulting in high rates of blindness and disability. Although several approaches exist in the treatment of RB, some children with the disease do not have satisfactory results because of various factors. Plant-derived natural products have shown definite therapeutic effects in the treatment of various tumors and are also widely used in the study of RB. We review plant-derived natural products used in the study of anti-RB to provide ideas for the clinical application of these drugs and the development of new therapeutic drugs.
Collapse
Affiliation(s)
- Jing-Chen Liu
- Department of Ophthalmology, Jiangxi Provincial Hospital of Integrated Traditional Chinese and Western Medicine, the Fourth Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330003, Jiangxi Province, China.,School of Clinical Medicine, Dali University, Dali 671000, Yunnan Province, China.,School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, Jiangxi Province, China
| | - Chun-Li Zhang
- Department of Ophthalmology, General Hospital of Southern Theatre Command, Guangzhou 510010, Guangdong Province, China
| | - Kai-Ye Dong
- Department of Ophthalmology, the First Affiliated Hospital of Dali University, Dali 671000, Yunnan Province, China
| | - Ming-Jun Li
- School of Clinical Medicine, Dali University, Dali 671000, Yunnan Province, China
| | - Shu-Guang Sun
- School of Clinical Medicine, Dali University, Dali 671000, Yunnan Province, China.,Department of Endocrinology, the First Affiliated Hospital of Dali University, Dali 671000, Yunnan Province, China
| | - Cai-Rui Li
- School of Clinical Medicine, Dali University, Dali 671000, Yunnan Province, China.,Department of Ophthalmology, the First Affiliated Hospital of Dali University, Dali 671000, Yunnan Province, China
| |
Collapse
|
26
|
Pei J, Cai L, Wang F, Xu C, Pei S, Guo H, Sun X, Chun J, Cong X, Zhu W, Zheng Z, Chen X. LPA 2 Contributes to Vascular Endothelium Homeostasis and Cardiac Remodeling After Myocardial Infarction. Circ Res 2022; 131:388-403. [PMID: 35920162 DOI: 10.1161/circresaha.122.321036] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Myocardial infarction (MI) is one of the most dangerous adverse cardiovascular events. Our previous study found that lysophosphatidic acid (LPA) is increased in human peripheral blood after MI, and LPA has a protective effect on the survival and proliferation of various cell types. However, the role of LPA and its receptors in MI is less understood. OBJECTIVES To study the unknown role of LPA and its receptors in heart during MI. METHODS AND RESULTS In this study, we found that mice also had elevated LPA level in peripheral blood, as well as increased cardiac expression of its receptor LPA2 in the early stages after MI. With adult and neonate MI models in global Lpar2 knockout (Lpar2-KO) mice, we found Lpar2 deficiency increased vascular leak leading to disruption of its homeostasis, so as to impaired heart function and increased early mortality. Histological examination revealed larger scar size, increased fibrosis, and reduced vascular density in the heart of Lpar2-KO mice. Furthermore, Lpar2-KO also attenuated blood flow recovery after femoral artery ligation with decreased vascular density in gastrocnemius. Our study revealed that Lpar2 was mainly expressed and altered in cardiac endothelial cells during MI, and use of endothelial-specific Lpar2 knockout mice phenocopied the global knockout mice. Additionally, adenovirus-Lpar2 and pharmacologically activated LPA2 significantly improved heart function, reduced scar size, increased vascular formation, and alleviated early mortality by maintaining vascular homeostasis owing to protecting vessels from leakage. Mechanistic studies demonstrated that LPA-LPA2 signaling could promote endothelial cell proliferation through PI3K-Akt/PLC-Raf1-Erk pathway and enhanced endothelial cell tube formation via PKD1-CD36 signaling. CONCLUSIONS Our results indicate that endothelial LPA-LPA2 signaling promotes angiogenesis and maintains vascular homeostasis, which is vital for restoring blood flow and repairing tissue function in ischemic injuries. Targeting LPA-LPA2 signal might have clinical therapeutic potential to protect the heart from ischemic injury.
Collapse
Affiliation(s)
- Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (J.P., Z.Z.)
| | - Lin Cai
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China (L.C.)
| | - Fang Wang
- State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuansheng Xu
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengqiang Pei
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongwei Guo
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Sun
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA (J.C.)
| | - Xiangfeng Cong
- State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weiquan Zhu
- Department of Medicine, Program in Molecular Medicine, Department of Internal Medicine, Division of Cardiovascular Medicine, Department of Pathology, University of Utah, Salt Lake City (W.Z.)
| | - Zhe Zheng
- Department of Cardiovascular Surgery (H.G., X.S., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China (J.P., Z.Z.)
| | - Xi Chen
- State Key Laboratory of Cardiovascular Disease (J.P., L.C., C.X., S.P., X.C., Z.Z.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Center of Laboratory Medicine (F.W., X. Cong, X. Chen), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
The FOXO family of transcription factors: key molecular players in gastric cancer. J Mol Med (Berl) 2022; 100:997-1015. [PMID: 35680690 DOI: 10.1007/s00109-022-02219-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 10/18/2022]
Abstract
Gastric cancer (GC) is the fifth most frequently diagnosed cancer worldwide and the third leading cause of cancer-related death with an oncological origin. Despite its decline in incidence and mortality in recent years, GC remains a global public problem that seriously threatens patients' health and lives. The forkhead box O proteins (FOXOs) are a family of evolutionarily conserved transcription factors (TFs) with crucial roles in cell fate decisions. In mammals, the FOXO family consists of four members FOXO1, 3a, 4, and 6. FOXOs play crucial roles in a variety of biological processes, such as development, metabolism, and stem cell maintenance, by regulating the expression of their target genes in space and time. An accumulating amount of evidence has shown that the dysregulation of FOXOs is involved in GC progression by affecting multiple cellular processes, including proliferation, apoptosis, invasion, metastasis, cell cycle progression, carcinogenesis, and resistance to chemotherapeutic drugs. In this review, we systematically summarize the recent findings on the regulatory mechanisms of FOXO family expression and activity and elucidate its roles in GC progression. Moreover, we also highlight the clinical implications of FOXOs in GC treatment.
Collapse
|
28
|
Yang L, Yue W, Zhang H, Zhang Z, Xue R, Dong C, Liu F, Chang N, Yang L, Li L. Dual Targeting of Angipoietin-1 and von Willebrand Factor by microRNA-671-5p Attenuates Liver Angiogenesis and Fibrosis. Hepatol Commun 2022; 6:1425-1442. [PMID: 35014213 PMCID: PMC9134804 DOI: 10.1002/hep4.1888] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/12/2021] [Accepted: 12/17/2021] [Indexed: 11/08/2022] Open
Abstract
Angipoietin-1 (Angpt1) and von Willebrand factor (VWF) are two important angiogenic molecules that can drive pathologic angiogenesis and progression of liver fibrosis in our previous study. MicroRNAs (miRs) participate in a variety of physiological and pathological processes, including angiogenesis. However, the critical miRs targeting Angpt1 or VWF and potential molecular mechanism underlying liver fibrosis-associated angiogenesis is not clear yet. Human liver tissues were obtained from patients with different chronic liver diseases. Mouse models of liver fibrosis were induced by injection of CCl4 or bile duct ligation (BDL) operation. MiR-671-5p was predicted to target Angpt1 and VWF from three databases (miRanda, RNA22v2, and miRwalk). MiR-671-5p expression was decreased in the fibrotic liver of human and mice, with a negative correlation with the levels of Angpt1, VWF, sphingosine kinase-1 (SphK1, the rate-limiting enzyme for sphingosine 1-phosphate [S1P] formation), transforming growth factor β1 (TGFβ1), hypoxia inducible factor (Hif)1α, Hif2α, and fibrosis markers. Importantly, miR-671-5p expression was down-regulated in fluorescence-activated cell sorted liver sinusoidal endothelial cells and hepatic stellate cells (HSCs) in CCl4 mice compared with control mice. In vitro miR-671-5p expression was also decreased in S1P-stimulated HSCs and TGFβ1-activated liver sinusoidal endothelial cells, negatively correlated with Angpt1 and VWF expression. MiR-671-5p directly targeted Angpt1 and VWF by luciferase reporter assays. In vivo administration of miR-671-5p agomir decreased the messenger RNA and protein levels of Anpgt1 and VWF, and attenuated CCl4 -induced or BDL-induced liver angiogenesis and fibrosis. Conclusion: We identify the negative regulation of miR-671-5p on Angpt1 and VWF and liver fibrosis-associated angiogenesis, which may provide promising targets for the prevention and treatment of liver disease.
Collapse
Affiliation(s)
- Le Yang
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Wenhui Yue
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Hang Zhang
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Zhi Zhang
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Renmin Xue
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Chengbin Dong
- Department of Interventional TherapyBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Fuquan Liu
- Department of Interventional TherapyBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Na Chang
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Lin Yang
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| | - Liying Li
- Department of Cell BiologyMunicipal Laboratory for Liver Protection and Regulation of RegenerationCapital Medical UniversityBeijingChina
| |
Collapse
|
29
|
Mechanism of N-Methyl-N-Nitroso-Urea-Induced Gastric Precancerous Lesions in Mice. JOURNAL OF ONCOLOGY 2022; 2022:3780854. [PMID: 35342404 PMCID: PMC8942688 DOI: 10.1155/2022/3780854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 11/17/2022]
Abstract
Early diagnosis and treatment of gastric precancerous lesions (GPL) are key factors for reducing the incidence and morbidity of gastric cancer. The study is aimed at examining GPL in mice induced by N-methyl-N-nitroso-urea (MNU) and to illustrate the underlying mechanisms of tumorigenesis. In this study, we utilized an in vivo MNU-induced GPL mouse model, and histopathological changes of the gastric mucosa were observed by hematoxylin and eosin (H&E-stain) and alcian blue (AB-PAS-stain). The level of miR-194-5p in the gastric mucosa was determined by real-time polymerase chain reaction. We used transmission electron microscopy to observe the effects of MNU on gastric chief cells and parietal cells. We performed immunohistochemical detection of HIF-1α, vWF, Ki-67, and P53, while the changes in the protein expression of key genes in LKB1-AMPK and AKT-FoxO3 signaling pathways were detected by western blot analysis. We demonstrated that the miR-194-5p expression was upregulated under hypoxia in GPL gastric tissues, and that a high miR-194-5p expression level closely related with tumorigenesis. Mechanistically, miR-194-5p exerted the acceleration of activities related to metabolic reprogramming through LKB1-AMPK and AKT-FoxO3 pathways. Furthermore, similar to miR-194-5p, high expression levels of AMPK and AKT were also related to the metabolic reprogramming of GPL. Moreover, we revealed the correlation between the expression levels of miR-194-5p, p-AMPKα, p-AKT, and FoxO3a. These findings suggest that miR-194-5p/FoxO3 pathway is important for the reversal of metabolic reprogramming in GPL. Thus, exploring strategies to regulate the miR-194-5p/FoxO3a pathway may provide an efficient strategy for the prevention and treatment of GPL.
Collapse
|
30
|
Zhu K, Deng C, Du P, Liu T, Piao J, Piao Y, Yang M, Chen L. G6PC indicated poor prognosis in cervical cancer and promoted cervical carcinogenesis in vitro and in vivo. Reprod Biol Endocrinol 2022; 20:50. [PMID: 35277194 PMCID: PMC8915493 DOI: 10.1186/s12958-022-00921-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/27/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The glucose-6-phosphatase catalytic subunit (G6PC) is a key enzyme that is involved in gluconeogenesis and glycogen decomposition during glycometabolism. Studies have shown that G6PC is abnormally expressed in various cancers and participates in the proliferation and metastasis of tumors. However, the role of G6PC in cervical cancer remains poorly established. METHODS To analyze the expression of G6PC in cervical cancer tissues in patients by immunohistochemistry. Effects of G6PC deregulation on cervical cancer phenotype were determined using MTT, colony formation, transwell, and wound-healing assays. And constructed a nude mouse xenograft tumor model and CAM assay in vivo. The effect of G6PC on glycolysis in cervical cancer was also evaluated. Effect of G6PC on PI3K/AKT/mTOR pathway was detected by Western blot assay. RESULTS In this study, G6PC expression was found to be upregulated in cervical cancer tissues, and this upregulated expression was associated with LN metastasis, clinical stage, recurrence, and disease-free survival and overall survival rates, indicating that G6PC could serve as a novel marker of early diagnosis in cervical cancer. G6PC promoted proliferation, invasion, epithelial mesenchymal transition (EMT) progression, and angiogenesis of cervical cancer cells. Mechanistically, G6PC activated PI3K/AKT/mTOR pathways. The PI3K/AKT pathway inhibitor, LY294002 could partially attenuate the effect. CONCLUSIONS G6PC plays a key role in the progression of cervical cancer, and overexpressed G6PC is closely related to patient LN metastasis, clinical stage, recurrence and shortened survival. G6PC promoted cervical cancer proliferation, invasion, migration, EMT progression, and angiogenesis, partially through activating the PI3K/AKT pathway. G6PC, as a metabolic gene, not only plays a role in metabolism, but also participates in the development of cervical cancer. Its complex metabolic and non metabolic effects may be a potential therapeutic target and worthy of further study.
Collapse
Affiliation(s)
- Kun Zhu
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
| | - Chunling Deng
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
| | - Pan Du
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
| | - Taorui Liu
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
| | - Junjie Piao
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Yingshi Piao
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China
| | - Meng Yang
- Department of Physiology, Medicine College, Jingchu University of Technology, Jingmen, 448000, China.
| | - Liyan Chen
- Cancer Research Center, Yanbian University Medical College, Gong Yuan Road No.977, Yanji, 133002, China.
- Key Laboratory of the Science and Technology Department of Jilin Province, Yanji, China.
| |
Collapse
|
31
|
Zhao L, Wang YF, Liu J, Jiang BH, Liu LZ. Human endothelial cells promote arsenic-transformed lung epithelial cells to induce tumor growth and angiogenesis through interleukin-8 induction. Aging (Albany NY) 2022; 14:2113-2130. [PMID: 35241635 PMCID: PMC8954972 DOI: 10.18632/aging.203930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/29/2022] [Indexed: 11/25/2022]
Abstract
Arsenic exposure is associated with lung cancer. Angiogenesis is essential for tumor development. However, the role and mechanism of human vascular endothelial cells in tumor growth and angiogenesis induced by arsenic-transformed bronchial epithelial (As-T) cells remain to be elucidated. In this study, we found that endothelial cells significantly increased As-T cell-induced tumor growth compared to those induced by As-T cells alone. To understand the molecular mechanism, we found that endothelial cells co-cultured with As-T cells or cultured in conditioned medium (CM) prepared from As-T cells showed much higher cell migration, proliferation, and tube formation compared to those co-cultured with BEAS-2B (B2B) cells or cultured in CM from B2B. We identified that higher levels of intracellular interleukin 8 (IL-8) were secreted by As-T cells, which activated IL-8/IL-8R signaling to promote endothelial cells migration and tube formation. IL-8 silencing and knockout (KO) in As-T cells, or IL-8 neutralizing antibody dramatically suppressed endothelial cell proliferation, migration, tube formation in vitro, and tumor growth and angiogenesis in vivo, suggesting a key role of IL-8 in As-T cells to induce angiogenesis via a paracrine effect. Finally, blocking of IL-8 receptors C-X-C chemokine receptor type 1 (CXCR1) and CXCR2 with neutralizing antibodies and chemical inhibitors inhibited tube formation, indicating that IL-8Rs on endothelial cells are necessary for As-T cell-induced angiogenesis. Overall, this study reveals an important molecular mechanism of arsenic-induced carcinogenesis, and suggests a new option to prevent and treat arsenic-induced angiogenesis.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yi-Fang Wang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jie Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bing-Hua Jiang
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ling-Zhi Liu
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
32
|
Eguchi R, Kawabe JI, Wakabayashi I. VEGF-Independent Angiogenic Factors: Beyond VEGF/VEGFR2 Signaling. J Vasc Res 2022; 59:78-89. [DOI: 10.1159/000521584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Tumors induce angiogenesis to acquire oxygen and nutrition from their adjacent microenvironment. Tumor angiogenesis has been believed to be induced primarily by the secretion of vascular endothelial growth factor-A (VEGF-A) from various tumors. VEGF-A binds to VEGF receptor 2 (VEGFR2), resulting in subsequent activation of cellular substances regulating cell proliferation, survival, and angiogenesis. Antiangiogenic therapies targeting the VEGF-A/VEGFR2 axis, including bevacizumab and ramucirumab, humanized monoclonal antibodies against VEGF-A and VEGFR2, respectively, have been proposed as a promising strategy aimed at preventing tumor growth, invasion, and metastasis. Phase III clinical trials using bevacizumab and ramucirumab have shown that not all tumor patients benefit from such antiangiogenic agents, and that some patients who initially benefit subsequently become less responsive to these antibodies, suggesting the possible existence of VEGF-independent angiogenic factors. In this review, we focus on VEGF-independent and VEGFR2-dependent tumor angiogenesis, as well as VEGFR2-independent tumor angiogenesis. Additionally, we discuss VEGF-independent angiogenic factors which have been reported in previous studies. Various molecular targeting drugs are currently being evaluated as potential antitumor therapies. We expect that precision medicine will permit the development of innovative antiangiogenic therapies targeting individual angiogenic factors selected on the basis of the genetic screening of tumors.
Collapse
|
33
|
Differential activation mechanisms of lipid GPCRs by lysophosphatidic acid and sphingosine 1-phosphate. Nat Commun 2022; 13:731. [PMID: 35136060 PMCID: PMC8826421 DOI: 10.1038/s41467-022-28417-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/21/2022] [Indexed: 12/23/2022] Open
Abstract
Lysophospholipids are bioactive lipids and can signal through G-protein-coupled receptors (GPCRs). The best studied lysophospholipids are lysophosphatidic acid (LPA) and sphingosine 1-phosphate (S1P). The mechanisms of lysophospholipid recognition by an active GPCR, and the activations of lysophospholipid GPCR–G-protein complexes remain unclear. Here we report single-particle cryo-EM structures of human S1P receptor 1 (S1P1) and heterotrimeric Gi complexes formed with bound S1P or the multiple sclerosis (MS) treatment drug Siponimod, as well as human LPA receptor 1 (LPA1) and Gi complexes in the presence of LPA. Our structural and functional data provide insights into how LPA and S1P adopt different conformations to interact with their cognate GPCRs, the selectivity of the homologous lipid GPCRs for S1P versus LPA, and the different activation mechanisms of these GPCRs by LPA and S1P. Our studies also reveal specific optimization strategies to improve the MS-treating S1P1-targeting drugs. Liu et al. report structures of human sphingosine 1-phosphate (S1P) receptor 1 (S1P1) in complex with Gi and S1P or the multiple sclerosis (MS) drug Siponimod, as well as human lysophosphatidic acid (LPA) receptor 1 (LPA1) in complex with Gi and LPA, revealing distinct conformations of the lysophospholipids interacting with their cognate GPCRs.
Collapse
|
34
|
Blogowski W, Dolegowska K, Deskur A, Dolegowska B, Starzynska T. Lipoxins and Resolvins in Patients With Pancreatic Cancer: A Preliminary Report. Front Oncol 2022; 11:757073. [PMID: 35087747 PMCID: PMC8787076 DOI: 10.3389/fonc.2021.757073] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Eicosanoids are bioactive lipids derived from arachidonic acid, which have emerged as key regulators of a wide variety of pathophysiological processes in recent times and are implicated as mediators of gastrointestinal cancer. In this study, we investigated the systemic levels of lipoxygenase (LOX)-derived lipoxin A4 and B4, together with resolvin D1 and D2 in patients with pancreatic adenocarcinoma (n = 68), as well as in healthy individuals (n = 32). Systemic concentrations of the aforementioned immunoresolvents were measured using an enzyme-linked immunosorbent assay (ELISA). In this study, we observed that compared with concentrations in healthy individuals, the peripheral concentrations of the aforementioned eicosanoids were significantly elevated (2- to 10-fold) in patients with pancreatic cancer (in all cases p<0.00001). No significant association was observed between eicosanoid levels and the TNM clinical staging. Furthermore, we observed no significant differences in concentrations of the analyzed bioactive lipids between patients diagnosed with early-stage (TNM stage I-II) and more advanced disease (TNM stage III-IV). Receiver operating characteristic (ROC) curve analysis of each aforementioned immunoresolvent showed area under the curve values ranging between 0.79 and 1.00. Sensitivity, specificity, as well as positive and negative predictive values of the eicosanoids involved in the detection/differentiation of pancreatic adenocarcinoma ranged between 56.8% and 100%. In summary, our research is the first study that provides clinical evidence to support a systemic imbalance in LOX-derived lipoxins and resolvins as the mechanism underlying the pathogenesis of pancreatic adenocarcinoma. This phenomenon occurs regardless of the clinical TNM stage of the disease. Furthermore, our study is the first to preliminarily highlight the role of peripheral levels of immunoresolvents, particularly resolvin D1, as potential novel biomarkers of pancreatic cancer in humans.
Collapse
Affiliation(s)
- Wojciech Blogowski
- Institute of Medical Sciences, University of Zielona Gora, Zielona Gora, Poland
| | - Katarzyna Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Anna Deskur
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Barbara Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Teresa Starzynska
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
35
|
Zhang W, Xing J, Liu T, Zhang J, Dai Z, Zhang H, Wang D, Tang D. Small extracellular vesicles: from mediating cancer cell metastasis to therapeutic value in pancreatic cancer. Cell Commun Signal 2022; 20:1. [PMID: 34980146 PMCID: PMC8722298 DOI: 10.1186/s12964-021-00806-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/20/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a highly malignant tumor and, is extremely difficult to diagnose and treat. Metastasis is one of the critical steps in the development of cancer and uses cell to cell communication to mediate changes in the microenvironment. Small extracellular vesicles (sEVs)-carry proteins, nucleic acids and other bioactive substances, and are important medium for communication between cells. There are two primary steps in sVEs-mediated metastasis: communication between pancreatic cancer cells and their surrounding microenvironment; and the communication between primary tumor cells and distant organ cells in distant organs that promotes angiogenesis, reshaping extracellular matrix, forming immunosuppressive environment and other ways to form appropriate pre-metastasis niche. Here, we explore the mechanism of localization and metastasis of pancreatic cancer and use sEVs as early biomarkers for the detection and treatment of pancreatic cancer. Video Abstract.
Collapse
Affiliation(s)
- Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Zhujiang Dai
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001 China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001 China
| |
Collapse
|
36
|
Liao J, Zheng Y, Hu M, Xu P, Lin L, Liu X, Wu Y, Huang B, Ye X, Li S, Duan R, Fu H, Huang J, Wen L, Fu Y, Kilby MD, Kenny LC, Baker PN, Qi H, Tong C. Impaired Sphingosine-1-Phosphate Synthesis Induces Preeclampsia by Deactivating Trophoblastic YAP (Yes-Associated Protein) Through S1PR2 (Sphingosine-1-Phosphate Receptor-2)-Induced Actin Polymerizations. Hypertension 2021; 79:399-412. [PMID: 34865521 DOI: 10.1161/hypertensionaha.121.18363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized trophoblast cells) cell invasion in a Hippo-signaling-dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA/ROCK induced actin polymerization. Mutation-based YAP-5SA demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.
Collapse
Affiliation(s)
- Jiujiang Liao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yangxi Zheng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Department of Biochemistry & Molecular Biology, University of Texas McGovern Medical School at Houston (Y.Z.).,Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX (Y.Z.)
| | - Mingyu Hu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ping Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Lin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xiyao Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Biao Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xuan Ye
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Sisi Li
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ran Duan
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Huijia Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Jiayu Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Wen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yong Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Mark D Kilby
- Institute of Metabolism and System Research, College of Medical & Dental Sciences, University of Birmingham and the Fetal Medicine Centre, Birmingham Women's and Children's Foundation Trust, United Kingdom (M.D.K.)
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, United Kingdom (L.C.K.)
| | - Philip N Baker
- College of Life Sciences, University of Leicester, United Kingdom (P.N.B.)
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Chongqing Women and Children's Health Center, China (H.Q.)
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| |
Collapse
|
37
|
Liu W, Luo H, Wei Q, Liu J, Wu J, Zhang Y, Chen L, Ren W, Shao L. Electrochemically derived nanographene oxide activates endothelial tip cells and promotes angiogenesis by binding endogenous lysophosphatidic acid. Bioact Mater 2021; 9:92-104. [PMID: 34820558 PMCID: PMC8586026 DOI: 10.1016/j.bioactmat.2021.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/08/2023] Open
Abstract
Graphene oxide (GO) exhibits good mechanical and physicochemical characteristics and has extensive application prospects in bone tissue engineering. However, its effect on angiogenesis is unclear, and its potential toxic effects are heavily disputed. Herein, we found that nanographene oxide (NGO) synthesized by one-step water electrolytic oxidation is smaller and shows superior biocompatibility. Moreover, NGO significantly enhanced angiogenesis in calvarial bone defect areas in vivo, providing a good microenvironment for bone regeneration. Endothelial tip cell differentiation is an important step in the initiation of angiogenesis. We verified that NGO activates endothelial tip cells by coupling with lysophosphatidic acid (LPA) in serum via strong hydrogen bonding interactions, which has not been reported. In addition, the mechanism by which NGO promotes angiogenesis was systematically studied. NGO-coupled LPA activates LPAR6 and facilitates the formation of migratory tip cells via Hippo/Yes-associated protein (YAP) independent of reactive oxygen species (ROS) stimulation or additional complex modifications. These results provide an effective strategy for the application of electrochemically derived NGO and more insight into NGO-mediated angiogenesis. Electrochemically derived nanographene oxide (NGO) has good cytocompatibility without upregulating reactive oxygen species. NGO exhibits better dispersibility and couples with endogenous lysophosphatidic acid (LPA) in body fluid. NGO enhances the angiogenesis by recruiting endogenous LPA and promoting endothelial tip cell formation.
Collapse
Affiliation(s)
- Wenjing Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Haiyun Luo
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Qinwei Wei
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Lili Chen
- Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wencai Ren
- Shenyang National Laboratory for Materials Science, Institute of Metal Research, Chinese Academy of Sciences, Shenyang, 110016, China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
- Corresponding author. Stomatological Hospital, Southern Medical University, Guangzhou 510280, China Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China.
| |
Collapse
|
38
|
Matas-Rico E, Frijlink E, van der Haar Àvila I, Menegakis A, van Zon M, Morris AJ, Koster J, Salgado-Polo F, de Kivit S, Lança T, Mazzocca A, Johnson Z, Haanen J, Schumacher TN, Perrakis A, Verbrugge I, van den Berg JH, Borst J, Moolenaar WH. Autotaxin impedes anti-tumor immunity by suppressing chemotaxis and tumor infiltration of CD8 + T cells. Cell Rep 2021; 37:110013. [PMID: 34788605 PMCID: PMC8761359 DOI: 10.1016/j.celrep.2021.110013] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 01/22/2023] Open
Abstract
Autotaxin (ATX; ENPP2) produces lysophosphatidic acid (LPA) that regulates multiple biological functions via cognate G protein-coupled receptors LPAR1–6. ATX/LPA promotes tumor cell migration and metastasis via LPAR1 and T cell motility via LPAR2, yet its actions in the tumor immune microenvironment remain unclear. Here, we show that ATX secreted by melanoma cells is chemorepulsive for tumor-infiltrating lymphocytes (TILs) and circulating CD8+ T cells ex vivo, with ATX functioning as an LPA-producing chaperone. Mechanistically, T cell repulsion predominantly involves Gα12/13-coupled LPAR6. Upon anti-cancer vaccination of tumor-bearing mice, ATX does not affect the induction of systemic T cell responses but, importantly, suppresses tumor infiltration of cytotoxic CD8+ T cells and thereby impairs tumor regression. Moreover, single-cell data from melanoma tumors are consistent with intratumoral ATX acting as a T cell repellent. These findings highlight an unexpected role for the pro-metastatic ATX-LPAR axis in suppressing CD8+ T cell infiltration to impede anti-tumor immunity, suggesting new therapeutic opportunities. Through LPA production, ATX modulates the tumor microenvironment in autocrine-paracrine manners. Matas-Rico et al. show that ATX/LPA is chemorepulsive for T cells with a dominant inhibitory role for Gα12/13-coupled LPAR6. Upon anticancer vaccination, tumor-intrinsic ATX suppresses the infiltration of CD8+ T cells without affecting their cytotoxic quality.
Collapse
Affiliation(s)
- Elisa Matas-Rico
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Elselien Frijlink
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Irene van der Haar Àvila
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Apostolos Menegakis
- Oncode Institute, Utrecht, the Netherlands; Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maaike van Zon
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Andrew J Morris
- Division of Cardiovascular Medicine, Gill Heart Institute and Lexington Veterans Affairs Medical Center, University of Kentucky, Lexington, KY, USA
| | - Jan Koster
- Laboratory for Experimental Oncology and Radiobiology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Fernando Salgado-Polo
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Sander de Kivit
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Telma Lança
- Oncode Institute, Utrecht, the Netherlands; Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Bari, Italy
| | - Zoë Johnson
- iOnctura SA, Campus Biotech Innovation Park, Geneva, Switzerland
| | - John Haanen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ton N Schumacher
- Oncode Institute, Utrecht, the Netherlands; Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anastassis Perrakis
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Inge Verbrugge
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Joost H van den Berg
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jannie Borst
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| | - Wouter H Moolenaar
- Division of Biochemistry, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
39
|
Dehghan M, Shahbazi S, Salehnia M. Effect of Lysophosphatidic Acid on the Vascular Endothelial Growth Factor Expression in Autotransplanted Mouse Ovaries Encapsulated in Sodium Alginate. J Family Reprod Health 2021; 15:91-98. [PMID: 34721597 PMCID: PMC8520664 DOI: 10.18502/jfrh.v15i2.6449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objective: The aim of this study was to evaluate the effect of lysophosphatidic acid (LPA) supplementation during in vitro culture and transplantation of mouse ovaries on the follicular development and expression of vascular endothelial growth factor (VEGF) as an angiogenesis factor at the mRNA and protein levels. Materials and methods: Three weeks old mice ovaries were cultured in the presence and absence of LPA for 24 hours, then they were capsulated in sodium alginate in the presence and absence of LPA as four experimental groups. After transplantation the vaginal smears were performed daily to evaluate the initiation of the estrous cycle. The morphology and follicular distribution were analyzed at the first and fourth estrous cycles using hematoxylin and eosin staining. Then in the groups that showed higher and lower follicular development the immunohistochemistry assay was conducted to identify VEGF protein expression, and the real time RT-PCR was done to analyze the expression of Vegf gene at the first estrus cycle. Results: The large size follicles and also the corpus luteum were prominent in all transplanted groups at fourth estrus cycle in comparison with intact control groups. The statistically lowest percentage of small size follicles and the highest percentages of large size follicles were seen in LPA+/LPA- group (p<0.05). The expression ratio of Vegf to β-actin was significantly higher in this group in comparison with non-LPA treated and intact control groups (p <0.05). Conclusion: LPA as an angiogenesis factor increases the follicular development in transplanted ovaries but it causes early discharge of ovarian reserve.
Collapse
Affiliation(s)
- Maryam Dehghan
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shirin Shahbazi
- Medical Genetic Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojdeh Salehnia
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
40
|
Biological functions and clinical significance of long noncoding RNAs in bladder cancer. Cell Death Discov 2021; 7:278. [PMID: 34611133 PMCID: PMC8492632 DOI: 10.1038/s41420-021-00665-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/02/2021] [Accepted: 09/17/2021] [Indexed: 12/24/2022] Open
Abstract
Bladder cancer (BCa) is one of the 10 most common cancers with high morbidity and mortality worldwide. Long noncoding RNAs (lncRNAs), a large class of noncoding RNA transcripts, consist of more than 200 nucleotides and play a significant role in the regulation of molecular interactions and cellular pathways during the occurrence and development of various cancers. In recent years, with the rapid advancement of high-throughput gene sequencing technology, several differentially expressed lncRNAs have been discovered in BCa, and their functions have been proven to have an impact on BCa development, such as cell growth and proliferation, metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, and drug-resistance. Furthermore, evidence suggests that lncRNAs are significantly associated with BCa patients' clinicopathological characteristics, especially tumor grade, TNM stage, and clinical progression stage. In addition, lncRNAs have the potential to more accurately predict BCa patient prognosis, suggesting their potential as diagnostic and prognostic biomarkers for BCa patients in the future. In this review, we briefly summarize and discuss recent research progress on BCa-associated lncRNAs, while focusing on their biological functions and mechanisms, clinical significance, and targeted therapy in BCa oncogenesis and malignant progression.
Collapse
|
41
|
Farahani DB, Akrami H, Moradi B, Mehdizadeh K, Fattahi MR. The Effect of hsa-miR-451b Knockdown on Biological Functions of Gastric Cancer Stem-Like Cells. Biochem Genet 2021; 59:1203-1224. [PMID: 33725258 DOI: 10.1007/s10528-021-10057-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 03/02/2021] [Indexed: 01/15/2023]
Abstract
Numerous researches have extensively studied factors such as microRNAs that lead to cancer. Thus, the current study's purpose is to investigate the biological consequences of hsa-miR-451b inhibition on the properties and functions of gastric cancer stem-like cells. First, gastric cancer stem-like cells were transfected by hsa-miR-451b inhibitor then we used real-time RT-PCR to evaluate its effect on the expression of hsa-miR-451b and two of its direct target genes, Stemness markers such as KLF4, SOX2, CD44, OCT3/4 and NANOG genes and finally Akt, PI3K, Bcl-2, Bax, CASP3 and PCNA genes involved in apoptosis. Here, we conducted a DNA Laddering assay to investigate apoptosis. The level of the MMP-2 and -9 Activities and Migration were examined by Zymography and Transwell invasion assay. HUVEC cells were used to investigate angiogenesis. The outcomes revealed that the level of the MMP-2 and -9 Activities, migration and angiogenesis decreased, but apoptosis was induced by inhibiting hsa-miR-451b. Evaluating KREMEN1 and CASK expression showed that the former increased, and the latter dropped under hsa-miR-451b inhibition. Also, upregulation of the KLF4 and SOX2 and downregulation of the CD44, OCT3/4, and NANOG decreased Self-renewal ability of gastric cancer stem cells under hsa-miR-451b inhibition. Even, under hsa-miR-451b inhibition, downregulation of Akt, PI3K, Bcl-2 and PCNA as well as upregulation of Bax and CASP3 revealed a movement towards apoptosis in MKN-45 stem-like cells. In summary, hsa-miR-451b is an oncomir in the carcinogenesis of gastric cancer stem-like cells and may be suggested as an appropriate therapeutic target for future gastric cancer treatment.
Collapse
Affiliation(s)
| | - Hassan Akrami
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, 71935-1311, Shiraz, Iran.
| | - Behrouz Moradi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Kiumars Mehdizadeh
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Mohammad Reza Fattahi
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, 71935-1311, Shiraz, Iran
| |
Collapse
|
42
|
Tan X, Fu J, Yuan Z, Zhu L, Fu L. ACNPD: The Database for Elucidating the Relationships Between Natural Products, Compounds, Molecular Mechanisms, and Cancer Types. Front Pharmacol 2021; 12:746067. [PMID: 34497528 PMCID: PMC8419280 DOI: 10.3389/fphar.2021.746067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/10/2021] [Indexed: 01/13/2023] Open
Abstract
Objectives: Cancer is well-known as a collection of diseases of uncontrolled proliferation of cells caused by mutated genes which are generated by external or internal factors. As the mechanisms of cancer have been constantly revealed, including cell cycle, proliferation, apoptosis and so on, a series of new emerging anti-cancer drugs acting on each stage have also been developed. It is worth noting that natural products are one of the important sources for the development of anti-cancer drugs. To the best of our knowledge, there is not any database summarizing the relationships between natural products, compounds, molecular mechanisms, and cancer types. Materials and methods: Based upon published literatures and other sources, we have constructed an anti-cancer natural product database (ACNPD) (http://www.acnpd-fu.com/). The database currently contains 521 compounds, which specifically refer to natural compounds derived from traditional Chinese medicine plants (derivatives are not considered herein). And, it includes 1,593 molecular mechanisms/signaling pathways, covering 10 common cancer types, such as breast cancer, lung cancer and cervical cancer. Results: Integrating existing data sources, we have obtained a large amount of information on natural anti-cancer products, including herbal sources, regulatory targets and signaling pathways. ACNPD is a valuable online resource that illustrates the complex pharmacological relationship between natural products and human cancers. Conclusion: In summary, ACNPD is crucial for better understanding of the relationships between traditional Chinese medicine (TCM) and cancer, which is not only conducive to expand the influence of TCM, but help to find more new anti-cancer drugs in the future.
Collapse
Affiliation(s)
- Xiaojie Tan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China.,MOE Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Jiahui Fu
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhaoxin Yuan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Lingjuan Zhu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Leilei Fu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
43
|
Stritt S, Koltowska K, Mäkinen T. Homeostatic maintenance of the lymphatic vasculature. Trends Mol Med 2021; 27:955-970. [PMID: 34332911 DOI: 10.1016/j.molmed.2021.07.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
The lymphatic vasculature is emerging as a multifaceted regulator of tissue homeostasis and regeneration. Lymphatic vessels drain fluid, macromolecules, and immune cells from peripheral tissues to lymph nodes (LNs) and the systemic circulation. Their recently uncovered functions extend beyond drainage and include direct modulation of adaptive immunity and paracrine regulation of organ growth. The developmental mechanisms controlling lymphatic vessel growth have been described with increasing precision. It is less clear how the essential functional features of lymphatic vessels are established and maintained. We discuss the mechanisms that maintain lymphatic vessel integrity in adult tissues and control vessel repair and regeneration. This knowledge is crucial for understanding the pathological vessel changes that contribute to disease, and provides an opportunity for therapy development.
Collapse
Affiliation(s)
- Simon Stritt
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Katarzyna Koltowska
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics, and Pathology, 751 85 Uppsala, Sweden.
| |
Collapse
|
44
|
Rudzitis-Auth J, Christoffel A, Menger MD, Laschke MW. Targeting sphingosine kinase-1 with the low MW inhibitor SKI-5C suppresses the development of endometriotic lesions in mice. Br J Pharmacol 2021; 178:4104-4118. [PMID: 34185874 DOI: 10.1111/bph.15601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/17/2021] [Accepted: 06/08/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND PURPOSE Limited evidence suggests that the sphingosine-1-phosphate/sphingosine kinase 1 (S1P/SPHK1) signalling pathway is involved in the pathogenesis of endometriosis. Therefore, we analyzed in this study whether the inhibition of SPHK1 and, consequently, decreased levels of S1P affected the vascularization and growth of endometriotic lesions. EXPERIMENTAL APPROACH Endometriotic lesions were surgically induced in the peritoneal cavity and the dorsal skinfold chamber of female BALB/c mice. The animals received a daily dose of the SPHK1 inhibitor SKI-5C or vehicle (control). Analyses involved the determination of lesion growth, cyst formation, microvessel density and cell proliferation within peritoneal endometriotic lesions by means of high-resolution ultrasound imaging, caliper measurement, histology and immunohistochemistry. In the dorsal skinfold chamber model the development of newly formed microvascular networks and their microhemodynamic parameters within endometriotic lesions were investigated by means of intravital fluorescence microscopy. KEY RESULTS SKI-5C significantly inhibited the development and vascularization of peritoneal endometriotic lesions, as indicated by a reduced growth and cyst formation, a lower microvessel density and a suppressed cell proliferation, when compared to vehicle-treated controls. Endometriotic lesions in dorsal skinfold chambers of SKI-5C-treated animals exhibited a significantly smaller lesion size, lower functional microvessel density, smaller microvessel diameters and a reduced blood perfusion of the newly developing microvascular networks. CONCLUSIONS AND IMPLICATIONS SPHK1/S1P signalling promotes the establishment and progression of endometriotic lesions. The inhibition of this pathway suppresses the development of endometriotic lesions, suggesting SPHK1 as a potential novel target for future endometriosis therapy.
Collapse
Affiliation(s)
| | - Anika Christoffel
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
45
|
Broadfield LA, Pane AA, Talebi A, Swinnen JV, Fendt SM. Lipid metabolism in cancer: New perspectives and emerging mechanisms. Dev Cell 2021; 56:1363-1393. [PMID: 33945792 DOI: 10.1016/j.devcel.2021.04.013] [Citation(s) in RCA: 346] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Tumors undergo metabolic transformations to sustain uncontrolled proliferation, avoid cell death, and seed in secondary organs. An increased focus on cancer lipid metabolism has unveiled a number of mechanisms that promote tumor growth and survival, many of which are independent of classical cellular bioenergetics. These mechanisms include modulation of ferroptotic-mediated cell death, support during tumor metastasis, and interactions with the cells of the tumor microenvironment. As such, targeting lipid metabolism for anti-cancer therapies is attractive, with recent work on small-molecule inhibitors identifying compounds to target lipid metabolism. Here, we discuss these topics and identify open questions.
Collapse
Affiliation(s)
- Lindsay A Broadfield
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Antonino Alejandro Pane
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ali Talebi
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute (LKI), KU Leuven, University of Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
46
|
Shahik SM, Salauddin A, Hossain MS, Noyon SH, Moin AT, Mizan S, Raza MT. Screening of novel alkaloid inhibitors for vascular endothelial growth factor in cancer cells: an integrated computational approach. Genomics Inform 2021; 19:e6. [PMID: 33840170 PMCID: PMC8042301 DOI: 10.5808/gi.20068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/11/2021] [Indexed: 12/26/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is expressed at elevated levels by most cancer cells, which can stimulate vascular endothelial cell growth, survival, proliferation as well as trigger angiogenesis modulated by VEGF and VEGFR (a tyrosine kinase receptor) signaling. The angiogenic effects of the VEGF family are thought to be primarily mediated through the interaction of VEGF with VEGFR-2. Targeting this signaling molecule and its receptor is a novel approach for blocking angiogenesis. In recent years virtual high throughput screening has emerged as a widely accepted powerful technique in the identification of novel and diverse leads. The high-resolution X-ray structure of VEGF has paved the way to introduce new small molecular inhibitors by structure-based virtual screening. In this study using different alkaloid molecules as potential novel inhibitors of VEGF, we proposed three alkaloid candidates for inhibiting VEGF and VEGFR mediated angiogenesis. As these three alkaloid compounds exhibited high scoring functions, which also highlights their high binding ability, it is evident that these alkaloids can be taken to further drug development pipelines for use as novel lead compounds to design new and effective drugs against cancer.
Collapse
Affiliation(s)
- Shah Md Shahik
- Molecular Biology Department, AFC Agro Biotech Ltd., Dhaka 1212, Bangladesh.,Bioinformatics Division, Disease Biology and Molecular Epidemiology Research Group (dBme), Chattogram 4202, Bangladesh
| | - Asma Salauddin
- Bioinformatics Division, Disease Biology and Molecular Epidemiology Research Group (dBme), Chattogram 4202, Bangladesh.,Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Md Shakhawat Hossain
- Bioinformatics Division, Disease Biology and Molecular Epidemiology Research Group (dBme), Chattogram 4202, Bangladesh.,Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Sajjad Hossain Noyon
- Bioinformatics Division, Disease Biology and Molecular Epidemiology Research Group (dBme), Chattogram 4202, Bangladesh.,Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Shagufta Mizan
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Md Thosif Raza
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| |
Collapse
|
47
|
Zhao WJ, Zhu LL, Yang WQ, Xu SJ, Chen J, Ding XF, Liang Y, Chen G. LPAR5 promotes thyroid carcinoma cell proliferation and migration by activating class IA PI3K catalytic subunit p110β. Cancer Sci 2021; 112:1624-1632. [PMID: 33540491 PMCID: PMC8019227 DOI: 10.1111/cas.14837] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Lysophosphatidic acid receptor 5 (LPAR5) is involved in mediating thyroid cancer progression, but the underlying mechanism needs to be further revealed. In this study, we confirmed that LPAR5 is upregulated in papillary thyroid carcinoma (PTC), especially in BRAF‐like PTC, by analyzing The Cancer Genome Atlas (TCGA) database and performing immunohistochemistry assay in human thyroid cancer tissues. LPAR5‐specific antagonist TC LPA5 4 treatment inhibited CGTH‐W3, TPC‐1, B‐CPAP, and BHT‐101 cell proliferation, CGTH‐W3 and TPC‐1 cell migration significantly. In vivo, TC LPA5 4 treatment could delay CGTH‐W3 xenograft growth in nude mice. We also found that LPAR5‐specific antagonist TC LPA5 4, PI3K inhibitor wortmannin, or mTOR inhibitor rapamycin pretreatment abrogated phosphorylation of Akt and p70S6K1 stimulated by LPA in CGTH‐W3 and TPC‐1 cells. Stimulating CGTH‐W3 cells transfected with pEGFPC1‐Grp1‐PH fusion protein with LPA resulted in the generation of phosphatidylinositol (3,4,5)‐triphosphate, which indicates that PI3K was activated by LPA directly. The p110β‐siRNA instead of p110α‐siRNA transfection abrogated the increase of levels of phosphorylated Akt and S6K1 stimulated by LPA. Furthermore, immunoprecipitation assay confirmed an interaction between LPAR5 and p110β. Overall, we provide new insights that the downregulation of LPAR5 decreased the proliferation and migration phenotype via the PI3K/Akt pathway. Inhibition of LPAR5 or the PI3K/Akt signal may be a novel therapeutic strategy for treating thyroid cancer.
Collapse
Affiliation(s)
- Wei-Jun Zhao
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China.,Graduate School of Medicine, Hebei North University, Zhangjiakou, China
| | - Liu-Lian Zhu
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China.,Graduate School of Medicine, Hebei North University, Zhangjiakou, China
| | - Wei-Qiang Yang
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China.,Graduate School of Medicine, Hebei North University, Zhangjiakou, China
| | - Shuai-Jun Xu
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China.,Graduate School of Medicine, Hebei North University, Zhangjiakou, China
| | - Jie Chen
- Department of Experimental and Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China
| | - Xiao-Fei Ding
- Department of Experimental and Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China
| | - Yong Liang
- Department of Clinical Medicine, School of Medicine, Taizhou University, Taizhou, China
| | - Guang Chen
- Department of Pharmacology, School of Medicine, Taizhou University, Taizhou, China
| |
Collapse
|
48
|
Zhao T, Xiong J, Chen W, Xu A, Zhu D, Liu J. Purification and Characterization of a Novel Fibrinolytic Enzyme from Cipangopaludina Cahayensis. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2805. [PMID: 34179197 PMCID: PMC8217531 DOI: 10.30498/ijb.2021.2805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Cipangopaludina cahayensis contains active fibrinolytic proteins and has been considered a potential anti-cancer agent.
However, its anti-cancer characteristics and functions have yet to be elucidated Objectives: To study the fibrinolytic activity and anticancer activity of crude protein extracts from Cipangopaludina cahayensis. Materials and Methods: Crude proteases were separated and extracted from the Cipangopaludina cahayensis through homogenization,
desalting, ammonium sulfate fractionation, dialysis, and ion exchange chromatography. The fibrinolytic activity
of extracted proteins was assessed using the fiber plate method. Total protein concentrations of the crude proteases
were determined via BCA assay. Molecular weights (MWs) were determined through SDS-PAGE electrophoresis. Results: The crude extract had a MW of ~ 50 kDa, and the highest protein concentration was 3.026 mg.mL-1.
The optimum pH for fibrinolytic activity was 7.0. Cell culture assays demonstrated that the addition of the
crude enzyme extracts to the human ovary cancer cell line Ovcar-3 resulted in significant growth defects. Conclusions: Our data showed that crude proteins purified from Cipangopaludina cahayensis are novel fibrinolytic proteases
and have potential anti-cancer propertie
Collapse
Affiliation(s)
- Tian Zhao
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Jinqi Xiong
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Wen Chen
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Ahui Xu
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Du Zhu
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| | - Jiantao Liu
- School of Life Sciences, Jiangxi Science & Technology Normal University, Nanchang 330013, China
| |
Collapse
|
49
|
Akhtar MF, Saleem A, Rasul A, Faran Ashraf Baig MM, Bin-Jumah M, Abdel Daim MM. Anticancer natural medicines: An overview of cell signaling and other targets of anticancer phytochemicals. Eur J Pharmacol 2020; 888:173488. [PMID: 32805253 DOI: 10.1016/j.ejphar.2020.173488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/23/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Therapies of cancer are as diverse as multifaceted the cancer is. Anticancer drugs include, but not limited to synthetic, semisynthetic and natural drugs and monoclonal antibodies. A recent decline in new drug development has led to the rebirth of herbal therapeutics in the form of dietary supplements and botanical preparations. Medicinal plants comprise of complex phytochemicals due to vast biosynthetic capacity. A wide array of phytochemicals has been pharmacologically evaluated for their chemo-preventive and chemotherapeutic potential for several decades. These phytochemicals target cancer at diverse sites such as apoptotic pathways, genetic and epigenetic mutations, damage to deoxyribonucleic acid, production of reactive oxygen species, autophagy, invasion and metastasis of cancer cells, and modulation of cell signaling through Janus-activated kinase/Signal transducer and activator of transcription, Notch, mitogen-activated protein kinase/Extracellular signal-regulated kinase, phosphatidylinositol 3-kinase/Protein kinase B/mammalian target of rapamycin, Nuclear factor kappa B, Wingless-related integration site and Transforming growth factor β pathways. This review focuses on the therapeutic targets of anticancer and chemo-preventive phytochemicals and their mode of action.
Collapse
Affiliation(s)
- Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan.
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - May Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel Daim
- Department of Zoology, College of Science, King Saud University, 2455, Riyadh, 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
50
|
Impacts of chitosan oligosaccharide (COS) on angiogenic activities. Microvasc Res 2020; 134:104114. [PMID: 33232706 DOI: 10.1016/j.mvr.2020.104114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 01/10/2023]
Abstract
It has been proved that chitosan oligosaccharide (COS) has a more favorable therapeutic applications such as wound healing and anti-tumor treatment, and can affect angiogenesis. For better understanding the effect of COS on angiogenic activities at cellular level, COS with different concentration and degree of polymerization (DP) were used to culture human umbilical vein endothelial cells (HUVECs) in this work. Cell proliferation activity, cell morphology, cell migration and angiogenesis associated factor expression of HUVECs were evaluated. The results indicated that COS at a high concentration of 400 μg/mL (COS(400)) and DP of 6 (Chitinhexaose Hydrochloride, COS6) had inhibitory effect on angiogenic activities of HUVECs. Specifically, COS(400) and COS6 inhibited cell proliferation activity, cell migration, and vascular endothelial cell growth factor (VEGF) expression of HUVECs. While COS at a low concentration (<400 μg/mL) and suitable polymerization degrees (DP < 6) had little significant effect on cell proliferation, migration, and VEGF expression of HUVECs, showing dose-dependent effect. These findings provided insight for the potential use of COS, for broadening its future applications in biomedical fields and functional materials area. It also helped guide the design and synthesis of chitosan-based materials as an angiogenesis inhibitor for anti-angiogenic therapy.
Collapse
|