1
|
Zhou J, Ma X, Liu X, Liu Y, Fu J, Qi Y, Liu H. The impact of histone lactylation on the tumor microenvironment and metabolic pathways and its potential in cancer therapy. Genes Genomics 2024; 46:991-1011. [PMID: 39127851 DOI: 10.1007/s13258-024-01554-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND The complexity of cancer is intricately linked to its multifaceted biological processes, including the roles of the tumor microenvironment (TME) as well as genetic and metabolic regulation. Histone lactylation has recently emerged as a novel epigenetic modification mechanism that plays a pivotal role in regulating cancer initiation, proliferation, invasion, and metastasis. OBJECTIVE This review aims to elucidate the role of histone lactylation in modulating various aspects of tumor biology, including DNA repair mechanisms, glycolytic metabolic abnormalities, functions of non-tumor cells in the TME, and the promotion of tumor inflammatory responses and immune escape. Additionally, the review explores potential therapeutic strategies targeting histone lactylation. METHODS A comprehensive literature review was performed, analyzing recent findings on histone lactylation and its impact on cancer biology. This involved a systematic examination of studies focusing on biochemical pathways, cellular interactions, and clinical implications related to histone lactylation. RESULTS Histone lactylation was identified as a critical regulator of tumor cell DNA repair mechanisms and glycolytic metabolic abnormalities. It also significantly influences the functions of non-tumor cells within the TME, promoting tumor inflammatory responses and immune escape. Moreover, histone lactylation acts as a multifunctional biological signaling molecule impacting immune responses within the TME. Various cell types within the TME, including T cells and macrophages, were found to regulate tumor growth and immune escape mechanisms through lactylation. CONCLUSION Histone lactylation offers a novel perspective on tumor metabolism and its role in cancer development. It presents promising opportunities for the development of innovative cancer therapies. This review underscores the potential of histone lactylation as a therapeutic target, paving the way for new strategies in cancer treatment.
Collapse
Affiliation(s)
- Juanhong Zhou
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Xinyun Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Xiaofeng Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Jiaojiao Fu
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yaling Qi
- The First Clinical Medical College of Gansu University of Chinese Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Huiling Liu
- Department of Obstetrics and Gynecology, Gansu Provincial Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
2
|
Lv L, Zhou F, Quan Y, Fan Y, Bao Y, Dou Y, Qu H, Dai X, Zhao H, Zheng S, Zhao C, Yang L. Demethylzeylasteral exerts potent efficacy against non-small-cell lung cancer via the P53 signaling pathway. Transl Oncol 2024; 46:101989. [PMID: 38781861 PMCID: PMC11141460 DOI: 10.1016/j.tranon.2024.101989] [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: 01/02/2024] [Revised: 04/18/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
Abstract
Lung cancer has one of the highest mortality rates worldwide, with non-small-cell lung cancer (NSCLC) constituting approximately 85% of all cases. Demethylzeylasteral (DEM), extracted from Tripterygium wilfordii Hook F, exhibits notable anti-tumor properties. In this study, we revealed that DEM could effectively induce NSCLC cell apoptosis. Specifically, DEM can dose-dependently suppress the viability and migration of human NSCLC cells. RNA-seq analysis revealed that DEM regulates the P53-signaling pathway, which was further validated by assessing crucial proteins involved in this pathway. Biacore analysis indicated that DEM has high affinity with the P53 protein. The CDX model demonstrated DEM's anti-tumor actions. This work provided evidence that DEM-P53 interaction stabilizes P53 protein and triggers downstream anti-tumor activities. These findings indicate that DEM treatment holds promise as a potential therapeutic approach for NSCLC, which warrants further clinical assessment in patients with NSCLC.
Collapse
Affiliation(s)
- Linxi Lv
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Feng Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Yizhou Quan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yiwei Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yunjia Bao
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yaning Dou
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyan Qu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuanxuan Dai
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Haiyang Zhao
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China
| | - Suqing Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Chengguang Zhao
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Lehe Yang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
3
|
Xu W, Tang Y, Yang Y, Wang C, Liu C, Zhang J, Zhao L, Wang G. Depletion of CPNE7 sensitizes colorectal cancer to 5-fluorouracil by downregulating ATG9B expression. J Cell Mol Med 2024; 28:e18261. [PMID: 38526029 PMCID: PMC10962129 DOI: 10.1111/jcmm.18261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/26/2024] Open
Abstract
We aimed to explore the biological function of CPNE7 and determine the impact of CPNE7 on chemotherapy resistance in colorectal cancer (CRC) patients. According to the Gene Expression Profiling Interactive Analysis database and previously published data, CPNE7 was identified as a potential oncogene in CRC. RT-qPCR and Western blotting were performed to verify the expression of CPNE7. Chi-square test was used to evaluate the associations between CPNE7 and clinical features. Cell proliferation, colony formation, cell migration and invasion, cell cycle and apoptosis were assessed to determine the effects of CPNE7. Transcriptome sequencing was used to identify potential downstream regulatory genes, and gene set enrichment analysis was performed to investigate downstream pathways. The effect of CPNE7 on 5-fluorouracil chemosensitivity was verified by half maximal inhibitory concentration (IC50). Subcutaneous tumorigenesis assay was used to examine the role of CPNE7 in sensitivity of CRC to chemotherapy in vivo. Transmission electron microscopy was used to detect autophagosomes. CPNE7 was highly expressed in CRC tissues, and its expression was correlated with T stage and tumour site. Knockdown of CPNE7 inhibited the proliferation and colony formation of CRC cells and promoted apoptosis. Knockdown of CPNE7 suppressed the expression of ATG9B and enhanced the sensitivity of CRC cells to 5-fluorouracil in vitro and in vivo. Knockdown of CPNE7 reversed the induction of the autophagy pathway by rapamycin and reduced the number of autophagosomes. Depletion of CPNE7 attenuated the malignant proliferation of CRC cells and enhanced the chemosensitivity of CRC cells to 5-fluorouracil.
Collapse
Affiliation(s)
- Weile Xu
- The Department of General surgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- The Department of General surgeryHebei Chest HospitalShijiazhuangHebeiChina
- The Second Department of SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Yujie Tang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Yang Yang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Changjing Wang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Chen Liu
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jianqing Zhang
- The Department of Gastrointestinal surgeryThe Third Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lianmei Zhao
- Scientific Research CenterThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Guiying Wang
- The Department of General surgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
- The Second Department of SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
4
|
韩 齐, 叶 梦, 金 齐. [Demethylzeylasteral inhibits proliferation, migration and invasion and promotes apoptosis of non-small cell lung cancer cells by inhibiting the AKT/CREB signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:280-288. [PMID: 38501413 PMCID: PMC10954516 DOI: 10.12122/j.issn.1673-4254.2024.02.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To investigate the mechanism underlying the inhibitory effects of Demethylzeylasteral (T-96) on non-small cell lung cancer (NSCLC) cells. METHODS We first examined the effects of different concentrations (1, 3, 10, and 30 μmol/L) of demethylzeylasteral on morphology and cell number of A549 and H1299 cells. The changes in proliferation, cell viability, migration, invasion, and apoptosis of A549 and H1299 cells following demethylzeylasteral treatment were detected using clone formation, CCK-8, cell scratch, Transwell, and flow cytometric assays, and the effect of SC79 treatment against demethylzeylasteral-induced cell apoptosis was assessed. Western blotting was performed to detect the changes in expressions of E-cadherin, N-cadherin, vimentin, Bax, Bcl-2 and cleaved caspase-3 and phosphorylation of AKT/CREB in demethylzeylasteral-treated A549 and H1299 cells and the cellular expressions of apoptotic proteins following treatment with both demethylzeylasteral and SC79. RESULTS T-96 treatment caused elongation of the cell body and widening of the intercellular space and significantly inhibited cell viability, proliferation, migration and invasion of A549 and H1299 cells (P < 0.05). Flow cytometry showed that demethylzeylasteral induced apoptosis in both A549 and H1299 cells, whereas SC79 treatment obviously attenuated its pro-apoptotic effect (P < 0.05). Western blotting revealed up-regulated expressions of Bax and cleaved caspase-3 proteins and lowered Bcl-2 expression level in demethylzeylasteral-treated A549 and H1299 cells, but cotreatment with SC79 obviously attenuated the expressions of the apoptotic proteins. T-96 significantly up-regulated the expression level of E-cadherin, down-regulated the expressions of N-cadherin and vimentin, and inhibited the phosphorylation of AKT and CREB in the two cell lines (P < 0.05). CONCLUSION T-96 inhibits the proliferation, migration and invasion and induces apoptosis of NSCLC cells possibly by inhibiting the AKT/CREB signaling pathway.
Collapse
Affiliation(s)
- 齐齐 韩
- 蚌埠医科大学检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, China
| | - 梦然 叶
- 蚌埠医科大学检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, China
| | - 齐力 金
- 蚌埠医科大学检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, China
- 蚌埠医科大学第二附属医院检验科,安徽 蚌埠 233080Department of Laboratory Medicine, Second Affiliated Hospital of Bengbu Medical University, Bengbu 233080, China
| |
Collapse
|
5
|
Li W, Han F, Tang K, Ding C, Xiong F, Xiao Y, Li C, Liang Q, Lee KY, Lee IS, Gao H. Inhibiting NF-κB-S100A11 signaling and targeting S100A11 for anticancer effects of demethylzeylasteral in human colon cancer. Biomed Pharmacother 2023; 168:115725. [PMID: 37879212 DOI: 10.1016/j.biopha.2023.115725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/27/2023] Open
Abstract
Colon cancer is a common and deadly malignancy of the gastrointestinal tract. Targeting proteins that inhibit tumor proliferation could lead to innovative treatment strategies for this disease. Demethylzeylasteral, extracted naturally from Tripterygium wilfordii Hook. f., demonstrates incredible anti-colon cancer activity. However, the molecular mechanism behind this requires further investigation. This study aims to identify crucial targets and mechanisms of demethylzeylasteral in treating colon cancer, making it a promising candidate for anti-tumor therapy. Through gene knockout, overexpression techniques, and double Luciferase experiments, we confirmed that demethylzeylasteral reduces S100A11 expression in HT29 cells and in vivo tumor models to anti-colon cancer. By conducting Surface Plasmon Resonance, immunofluorescence staining, and confocal laser microscopy observations, we verified the direct interaction between demethylzeylasteral and S100A11, and explored the impact of S100A11's subcellular localization on cell proliferation. Demethylzeylasteral inhibited S100A11 expression and exhibited anti-cancer activity in both in vitro and in vivo colon cancer models. Conversely, overexpression of S100A11 hindered apoptosis induced by demethylzeylasteral. Additionally, we found that knockdown or overexpression of NF-κB respectively decreased or increased S100A11 expression, subsequently affecting cell proliferation. The dual Luciferase reporting experiment revealed that NF-κB is an upstream transcription factor regulating S100A11 expression. And Surface plasmon resonance confirmed that S100A11 can directly interact with demethylzeylasteral, this interaction limited the transport of S100A11 from the cytoplasm to nucleus, attenuation S100A11 mediated cell proliferation effect.
Collapse
Affiliation(s)
- Wenqing Li
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China; College of Pharmacy, Chonnam National University, Gwangju 61186, the Republic of Korea
| | - Fubo Han
- College of Pharmacy, Chonnam National University, Gwangju 61186, the Republic of Korea
| | - Kaifan Tang
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Chengjie Ding
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Fen Xiong
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yina Xiao
- College of Pharmacy, Chonnam National University, Gwangju 61186, the Republic of Korea
| | - Chen Li
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Qian Liang
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Kwang Youl Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, the Republic of Korea.
| | - Ik-Soo Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, the Republic of Korea.
| | - Hongchang Gao
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325035, PR China.
| |
Collapse
|
6
|
Cicchi C, Paoli P, Modesti A, Mannelli F, Scicutella F, Buccioni A, Fontanarosa C, Luti S, Pazzagli L. Effect of Bovine Milk Peptides on Cell Inflammation, Proliferation and Differentiation: Milk Potential Benefits Are Preserved in an Unconventional Cow Feeding Strategy. BIOLOGY 2023; 12:1162. [PMID: 37759562 PMCID: PMC10525111 DOI: 10.3390/biology12091162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
Animal feeding through the reuse of agro-industrial by-products in one of the ultimate goals of sustainable agriculture. Olive oil pomace (OOP) produced as a waste product during olive oil milling has been used as an ingredient in the diet for Holstein lactating cows. Recent findings have shown no decrease in animal performance, feed intake or detrimental effect on rumen microbiota. In contrast, an improvement in C18 polyunsaturated fatty acids has been observed. In this work, the milk protein content from cows fed a commercial diet (CON) or an experimental one supplemented with OOP was determined and compared, and the peptides derived from the simulated gastrointestinal digestion of raw milk were analyzed. After fractionation via RP-HPLC, peptides were characterized for their biological activity on different cell lines. The ability to reduce both the intracellular ROS content and the expression of inflammatory markers, such as Cyclooxygenase, isoenzyme 2 (COX-2) and inducible Nitric Oxide Synthase (iNOS), as well as the remarkable properties to induce cell differentiation and to slow down the proliferation of human intestinal cancer cells, enable us to define them as bioactive peptides. In spite of there being no observed significant difference between the healthy activity of CON and OOP peptides, the results allow us to broaden the knowledge about the biological activity of these bioactive peptides and to confirm that agro-industrial by-products may be successfully incorporated into the feeding strategy of dairy cows.
Collapse
Affiliation(s)
- Costanza Cicchi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (C.C.); (P.P.); (A.M.); (L.P.)
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (C.C.); (P.P.); (A.M.); (L.P.)
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (C.C.); (P.P.); (A.M.); (L.P.)
| | - Federica Mannelli
- Department of Agricultural, Food, Environmental and Forestry Sciences and Technologies, University of Florence, Piazzale delle Cascine 18, 50144 Florence, Italy; (F.M.); (F.S.); (A.B.)
| | - Federica Scicutella
- Department of Agricultural, Food, Environmental and Forestry Sciences and Technologies, University of Florence, Piazzale delle Cascine 18, 50144 Florence, Italy; (F.M.); (F.S.); (A.B.)
| | - Arianna Buccioni
- Department of Agricultural, Food, Environmental and Forestry Sciences and Technologies, University of Florence, Piazzale delle Cascine 18, 50144 Florence, Italy; (F.M.); (F.S.); (A.B.)
| | - Carolina Fontanarosa
- Department of Chemical Sciences, University of Naples Federico II, 80138 Naples, Italy;
- Consorzio Interuniversitario I.N.B.B., Viale Medaglie D’Oro, 00136 Rome, Italy
| | - Simone Luti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (C.C.); (P.P.); (A.M.); (L.P.)
| | - Luigia Pazzagli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (C.C.); (P.P.); (A.M.); (L.P.)
| |
Collapse
|
7
|
Chang Y, Kang P, Cui T, Guo W, Zhang W, Du P, Yi X, Guo S, Gao T, Li C, Li S. Pharmacological inhibition of demethylzeylasteral on JAK-STAT signaling ameliorates vitiligo. J Transl Med 2023; 21:434. [PMID: 37403086 DOI: 10.1186/s12967-023-04293-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND The activation of CD8+ T cells and their trafficking to the skin through JAK-STAT signaling play a central role in the development of vitiligo. Thus, targeting this key disease pathway with innovative drugs is an effective strategy for treating vitiligo. Natural products isolated from medicinal herbs are a useful source of novel therapeutics. Demethylzeylasteral (T-96), extracted from Tripterygium wilfordii Hook F, possesses immunosuppressive and anti-inflammatory properties. METHODS The efficacy of T-96 was tested in our mouse model of vitiligo, and the numbers of CD8+ T cells infiltration and melanocytes remaining in the epidermis were quantified using whole-mount tail staining. Immune regulation of T-96 in CD8+ T cells was evaluated using flow cytometry. Pull-down assay, mass spectrum analysis, molecular docking, knockdown and overexpression approaches were utilized to identify the target proteins of T-96 in CD8+ T cells and keratinocytes. RESULTS Here, we found that T-96 reduced CD8+ T cell infiltration in the epidermis using whole-mount tail staining and alleviated the extent of depigmentation to a comparable degree of tofacitinib (Tofa) in our vitiligo mouse model. In vitro, T-96 decreased the proliferation, CD69 membrane expression, and IFN-γ, granzyme B, (GzmB), and perforin (PRF) levels in CD8+ T cells isolated from patients with vitiligo. Pull-down assays combined with mass spectrum analysis and molecular docking showed that T-96 interacted with JAK3 in CD8+ T cell lysates. Furthermore, T-96 reduced JAK3 and STAT5 phosphorylation following IL-2 treatment. T-96 could not further reduce IFN-γ, GzmB and PRF expression following JAK3 knockdown or inhibit increased immune effectors expression upon JAK3 overexpression. Additionally, T-96 interacted with JAK2 in IFN-γ-stimulated keratinocytes, inhibiting the activation of JAK2, decreasing the total and phosphorylated protein levels of STAT1, and reducing the production and secretion of CXCL9 and CXCL10. T-96 did not significantly inhibit STAT1 and CXCL9/10 expression following JAK2 knockdown, nor did it suppress upregulated STAT1-CXCL9/10 signaling upon JAK2 overexpression. Finally, T-96 reduced the membrane expression of CXCR3, and the culture supernatants pretreated with T-96 under IFN-γ stressed keratinocytes markedly blocked the migration of CXCR3+CD8+ T cells, similarly to Tofa in vitro. CONCLUSION Our findings demonstrated that T-96 might have positive therapeutic responses to vitiligo by pharmacologically inhibiting the effector functions and skin trafficking of CD8+ T cells through JAK-STAT signaling.
Collapse
Affiliation(s)
- Yuqian Chang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Pan Kang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Tingting Cui
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Weinan Guo
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Weigang Zhang
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Pengran Du
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Xiuli Yi
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China.
| | - Shuli Li
- Department of Dermatology, Fourth Military Medical University, Xijing Hospital, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
8
|
Vijayan N, Perumal MK. A critical review on anti-fibrotic phytochemicals targeting activated hepatic stellate cells. J Food Biochem 2022; 46:e14438. [PMID: 36209494 DOI: 10.1111/jfbc.14438] [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: 07/13/2022] [Revised: 08/29/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is a major health concern occurring worldwide. It arises due to prolonged wound healing response of various insults like viral, autoimmune, cholestatic, drug-induced, and metabolic diseases. Currently, there is no clinically approved drug for liver fibrosis treatment. Hepatic stellate cells are the principal liver cells that are activated during liver fibrosis, and targeting these activated cells is an ideal therapeutic strategy. Numerous phytochemicals have been demonstrated in vitro and in vivo treating experimental liver fibrosis; however, none of them have been clinically approved for therapeutic use. This review mainly focuses on such hepatoprotective phytochemicals reported inhibiting major signaling pathways that are dysregulated in activated hepatic stellate cells. PRACTICAL APPLICATIONS: Liver fibrosis is a global health concern and there is no FDA approved drug to treat liver fibrosis. Although notable pharmacological agents like pentoxifylline, gliotoxin, imatinibmesylate, Gleevec, and so on are reported to exhibit anti-fibrotic effect, the major concern is their side effect. Hence, phytochemicals are promising candidates that could be employed against liver fibrosis. In this review, the anti-fibrotic potential of phytochemicals targeting activated HSCs are summarized. Understanding these phytochemicals will further help in the development of agents that are more effective against liver fibrosis.
Collapse
Affiliation(s)
- Nivya Vijayan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Madan Kumar Perumal
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
9
|
Sun X, Shen B, Yu H, Wu W, Sheng R, Fang Y, Guo R. Therapeutic potential of demethylzeylasteral, a triterpenoid of the genus Tripterygium wilfordii. Fitoterapia 2022; 163:105333. [DOI: 10.1016/j.fitote.2022.105333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/28/2022]
|
10
|
Chemopreventive Effect on Human Colon Adenocarcinoma Cells of Styrylquinolines: Synthesis, Cytotoxicity, Proapoptotic Effect and Molecular Docking Analysis. Molecules 2022; 27:molecules27207108. [PMID: 36296703 PMCID: PMC9607578 DOI: 10.3390/molecules27207108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
Seven styrylquinolines were synthesized in this study. Two of these styrylquinolines are new and were elucidated by spectroscopic analysis. The chemopreventive potential of these compounds was evaluated against SW480 human colon adenocarcinoma cells, its metastatic derivative SW620, and normal cells (HaCaT). According to the results, compounds 3a and 3d showed antiproliferative activity in SW480 and SW620 cells, but their effect seemed to be caused by different mechanisms of action. Compound 3a induced apoptosis independent of ROS production, as evidenced by increased levels of caspase 3, and had an immunomodulatory effect, positively regulating the production of different immunological markers in malignant cell lines. In contrast, compound 3d generated a pro-oxidant response and inhibited the growth of cancer cells, probably by another type of cell death other than apoptosis. Molecular docking studies indicated that the most active compound, 3a, could efficiently bind to the proapoptotic human caspases-3 protein, a result that could provide valuable information on the biochemical mechanism for the in vitro cytotoxic response of this compound in SW620 colon carcinoma cell lines. The obtained results suggest that these compounds have chemopreventive potential against CRC, but more studies should be carried out to elucidate the molecular mechanisms of action of each of them in depth.
Collapse
|
11
|
Xu L, Zhao B, Yang L, Dong X, Yang X, Mao Y. Demethylzeylasteral reduces the level of proteinuria in diabetic nephropathy: Screening of network pharmacology and verification by animal experiment. Pharmacol Res Perspect 2022; 10:e00976. [PMID: 35716119 PMCID: PMC9206407 DOI: 10.1002/prp2.976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 11/08/2022] Open
Abstract
This study aimed to use network pharmacology to detail the natural components isolated from Triptergium wilfordii Hook F (TwHF) and examine the effect of the main component (demethylzeylasteral, DEM) on rat models of diabetic nephropathy (DN). In this study, we used network pharmacology to detail the natural components isolated from TwHF, referenced a gene library when screening for components effective in the management of DN, and DEM was confirmed in DN rats. All data were analyzed using the Discovery Studio 4.5 System and the systems Dock online docking method platform. All 24 rats were divided into 4 groups: control, DN, TwHF, and DEM. Blood and urine samples were tested at 0, 8, and 12 weeks. Renal histopathological changes were scored. Network pharmacology indicated that 370 compounds and 46 small molecules (including DEM) were biologically active constituents of TwHF, mainly affecting the inflammatory response through PI3K-Akt and Jak-STAT pathways. Proteinuria in the TwHF and DEM groups was significantly lower than in the DN group (p ≤ .001), and the decrease in proteinuria in the DEM group was more obvious than in the TwHF group (p = .004). The tubular interstitial scores were better in the DEM group than in the TwHF and DN groups. These results indicate that DEM effectively reduced proteinuria and alleviated the tubular interstitial changes in rat models of DN, which may be provide a scientific foundation for the development of novel drugs for treatment of DN.
Collapse
Affiliation(s)
- Lengnan Xu
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingP.R. China
| | - Ban Zhao
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingP.R. China
| | - Liping Yang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingP.R. China
| | - Xinyi Dong
- Tianjin Medical University Eye HospitalTianjinP.R. China
| | - Xue Yang
- Department of Pharmacy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingP.R. China
| | - Yonghui Mao
- Department of Nephrology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingP.R. China
| |
Collapse
|
12
|
Pan L, Feng F, Wu J, Fan S, Han J, Wang S, Yang L, Liu W, Wang C, Xu K. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res 2022; 181:106270. [PMID: 35605812 DOI: 10.1016/j.phrs.2022.106270] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/04/2022] [Accepted: 05/18/2022] [Indexed: 12/12/2022]
Abstract
Cancer stem cells drive tumor initiation, progression, and recurrence, which compromise the effectiveness of anti-tumor drugs. Here, we report that demethylzeylasteral (DML), a triterpene anti-tumor compound, suppressed tumorigenesis of liver cancer stem cells (LCSCs) by interfering with lactylation of a metabolic stress-related histone. Using RNA sequencing (RNA-seq) and gas chromatography-mass spectrometric (GC-MS) analysis, we showed that the glycolysis metabolic pathway contributed to the anti-tumor effects of DML, and then focused on lactate downstream regulation as the molecular target. Mechanistically, DML opposed the progress of hepatocellular carcinoma (HCC), which was efficiently facilitated by the increase in H3 histone lactylation. Two histone modification sites: H3K9la and H3K56la, which were found to promote tumorigenesis, were inhibited by DML. In addition, we used a nude mouse tumor xenograft model to confirm that the anti-liver cancer effects of DML are mediated by regulating H3 lactylation in vivo. Our findings demonstrate that DML suppresses the tumorigenicity induced by LCSCs by inhibiting H3 histone lactylation, thus implicating DML as a potential candidate for the supplementary treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Lianhong Pan
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing 400030, China
| | - Fan Feng
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Jiaqin Wu
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Shibing Fan
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, China
| | - Juanjuan Han
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Shunxi Wang
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Li Yang
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Wanqian Liu
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| | - Chunli Wang
- National Innovation and Attracting Talents "111" base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| | - Kang Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China.
| |
Collapse
|
13
|
Tabnak P, Masrouri S, Mafakheri A. Natural products in suppressing glioma progression: A focus on the role of microRNAs. Phytother Res 2022; 36:1576-1599. [PMID: 35174549 DOI: 10.1002/ptr.7414] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/07/2021] [Accepted: 01/29/2022] [Indexed: 11/06/2022]
Abstract
Glioma is one of the most common malignancies of the central nervous system. Due to inadequate response to the current treatments available, glioma has been at the center of recent cancer studies searching for novel treatment strategies. This has prompted an intensive search using linkage studies and preliminary evidence to gain efficient insight into the mechanisms involved in the alleviation of the pathogenesis of glioma mediated by miRNAs, a group of noncoding RNAs that affect gene expression posttranscriptionally. Dysregulated expression of miRNAs can exacerbate the malignant features of tumor cells in glioma and other cancers. Natural products can exert anticancer effects on glioma cells by stimulating the expression levels of tumor suppressor miRNAs and repressing the expression levels of oncogenic miRNAs. In this review, we aimed to collect and analyze the literature addressing the roles of natural products in the treatment of glioma, with an emphasis on their involvement in the regulation of miRNAs.
Collapse
Affiliation(s)
- Peyman Tabnak
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Masrouri
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asrin Mafakheri
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
14
|
Li Y, Su Y, Zhao Y, Hu X, Zhao G, He J, Wan S, Lü M, Cui H. Demethylzeylasteral inhibits proliferation, migration, and invasion through FBXW7/c-Myc axis in gastric cancer. MedComm (Beijing) 2021; 2:467-480. [PMID: 34766156 PMCID: PMC8554662 DOI: 10.1002/mco2.73] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is one of the most familiar malignancy in the digestive system. Demethylzeylasteral (Dem), a natural functional monomer extracted from Tripterygium wilfordii Hook F, shows anti‐tumor effects in a variety of cancers, including GC, however, with the underlying mechanism poorly understood. In our study, we show that Dem inhibits the proliferation, migration, and invasion of GC cells, which are mediated by down‐regulating c‐Myc protein levels. Mechanistically, Dem reduces the stability of c‐Myc by up‐regulating FBXW7, an E3 ubiquitin ligase. Moreover, in xenograft tumor model experiment, Dem also inhibits GC, which depends on suppressing c‐Myc expression. Finally, Dem enhances GC cell chemosensitivity to the combination treatment of 5‐Fluorouracil (5‐Fu) and doxorubicin (DOX) in vitro. Together, Dem exerts anti‐neoplastic activities through destabilizing and suppressing c‐Myc, establishing a theory foundation for using it in future treatment of GC.
Collapse
Affiliation(s)
- Yongsen Li
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Yongyue Su
- Department of Orthopaedic 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army Kunming China
| | - Yuzu Zhao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Xiaosong Hu
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Gaichao Zhao
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Jiang He
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Sicheng Wan
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China
| | - Muhan Lü
- Department of Gastroenterology The Affiliated Hospital of Southwest Medical University Luzhou China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology College of Sericulture Textile and Biomass sciences Southwest University Chongqing China.,Department of Gastroenterology The Affiliated Hospital of Southwest Medical University Luzhou China.,Cancer Centre Medical Research Institute Southwest University Chongqing China
| |
Collapse
|