1
|
Chen G, Zhang C, Zou J, Zhou Z, Zhang J, Yan Y, Liang Y, Tang G, Chen G, Xu X, Wang N, Feng Y. Coptidis rhizoma and berberine as anti-cancer drugs: A 10-year updates and future perspectives. Pharmacol Res 2025; 216:107742. [PMID: 40258505 DOI: 10.1016/j.phrs.2025.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025]
Abstract
Cancer continues to be among the most substantial health challenges globally. Among various natural compounds, berberine, an isoquinoline alkaloid obtained from Coptidis Rhizoma, has garnered considerable attention for its broad-spectrum biological activities, including anti-inflammatory, antioxidant, anti-diabetic, anti-obesity, and anti-microbial activities. Furthermore, berberine exhibits a broad spectrum of anti-cancer efficacy against various malignancies, such as ovarian, breast, lung, gastric, hepatic, colorectal, cervical, and prostate cancers. Its anti-cancer mechanisms are multifaceted, encompassing the inhibition of cancer cell proliferation, the prevention of metastasis, the induction of apoptosis, the facilitation of autophagy, the modulation of the tumor microenvironment and gut microbiota, and the enhancement of the efficacy of conventional therapeutic strategies. This paper offers an exhaustive overview of the cancer-fighting characteristics of Coptidis Rhizoma and berberine, while also exploring recent developments in nanotechnology aimed at enhancing the bioavailability of berberine. Furthermore, the side effects and safety of berberine are addressed as well. The potential role of artificial intelligence in optimizing berberine's therapeutic applications is also highlighted. This paper provides precious perspectives on the prospective application of Coptidis Rhizoma and berberine in the prevention and management of cancer.
Collapse
Affiliation(s)
- Guoming Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Jiayi Zou
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zitian Zhou
- The Fourth School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiayi Zhang
- The First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Yan
- The School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yinglan Liang
- The Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guoyi Tang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Guang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Xiaoyu Xu
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong.
| |
Collapse
|
2
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [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: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
3
|
Ning S, Chen Y, Zhu H. LINC00968 accelerates osteogenic differentiation of bone marrow mesenchymal stem cells via the miR-17-5p/STAT3 axis. J Orthop Surg Res 2025; 20:242. [PMID: 40050898 PMCID: PMC11883945 DOI: 10.1186/s13018-025-05627-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND BMSCs with robust osteogenic differentiation capacity can participate in the repair of osteoporotic (OP) bone. Long non-coding RNAs (LncRNAs) serve as crucial regulators of osteogenic differentiation. This study aims to investigate the clinical implications of LINC00968 in OP and elucidate its molecular mechanisms. METHODS Patients with OP and controls without OP were enrolled. RT-qPCR was utilized the quantify the levels of LINC00968, miR-17-5p, STAT3, and osteogenic differentiation markers. ROC curve was conducted to evaluate the diagnostic significance. Osteogenic differentiation medium (OM) induced hBMSCs. Flow cytometry was used to examine apoptosis. DLR and RIP assay were performed to validate target binding. RESULTS LINC00968 was notably decreased in the serum and bone tissue of patients with OP, whereas it was markedly elevated during osteogenic differentiation of hBMSCs. LINC00968 has 78.65% sensitivity and 71.95% specificity in identifying OP patients from controls. Silencing of LINC00968 sharply diminished ALP activity and osteogenic differentiation markers levels while promoting apoptosis in hBMSCs under OM induction. However, this reduction was notably reversed by the administration of a miR-17-5p inhibitor. Molecularly, miR-17-5p directly targets LINC00968 and STAT3. CONCLUSIONS Our results indicate that LINC00968 downregulation is a diagnostic biomarker for OP, facilitating osteogenic differentiation and inhibiting apoptosis via miR-17-5p/STAT3 axis, suggesting a new therapeutic target for OP progression.
Collapse
Affiliation(s)
- Shanglong Ning
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yang Chen
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Hui Zhu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, No.1, Huanhu West Road, North Sports Institute Road, Hexi District, Tianjin, 300000, China.
| |
Collapse
|
4
|
Xiang L, Shen Y, Liu S, Fan B, Zhan J, Zhou Y, Jiang B, Wang M, Liu Q, Liu X, Zou Y, Sun S. Guggulsterone ameliorates psoriasis by inhibiting keratinocyte proliferation and inflammation through induction of miR-17 directly targeting JAK1 and STAT3. Biochem Pharmacol 2025; 233:116745. [PMID: 39793717 DOI: 10.1016/j.bcp.2025.116745] [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: 08/17/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
The pathogenesis of psoriasis involves hyperproliferation of epidermal keratinocytes and abnormal interactions between activated keratinocytes and infiltrating immune cells. Emerging evidence has shown that keratinocytes play essential roles in both the initiation and maintenance of psoriasis, suggesting that exposing keratinocytes to agents with antiproliferative and anti-inflammatory effects may be effective for psoriasis treatment. Guggulsterone (GS), a plant sterol derived from the gum resin of Commiphora wightii, possesses a variety of pharmacological activities. However, the effects of GS on psoriasis and the underlying mechanism have not been elucidated. In this study, we evaluated the therapeutic effect of GS on psoriasis using an imiquimod-induced psoriasis mouse model and investigated the effect of GS on human keratinocytes and the underlying mechanism. We found that GS effectively alleviated psoriasis-like skin lesions in imiquimod-induced psoriasis model mice and that GS suppressed the proliferation, migration, and production of proinflammatory cytokines, chemokines and antimicrobial peptides in keratinocytes. Transcriptome analysis by RNA-seq revealed that the differentially expressed genes (DEGs) induced by GS in keratinocytes were intricately linked to the pathogenesis of psoriasis. Furthermore, STAT3, a key player in the development and pathogenesis of psoriasis, was identified as a critical downstream mediator of GS in keratinocytes. Mechanistically, GS upregulated the expression of miR-17-5p, which directly binds to the 3'-untranslated regions (3'UTRs) of JAK1 and STAT3, leading to the downregulation of JAK1 and STAT3 expression. Collectively, these findings suggest that GS may serve as an effective natural compound for the treatment of psoriasis.
Collapse
Affiliation(s)
- Lu Xiang
- Department of Dermatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan 250011, China; The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yangli Shen
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Shuangteng Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Bowen Fan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Jiafeng Zhan
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Yadi Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China
| | - Xiaofei Liu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Genetics, School of Basic Medical Sciences, Shandong University, Jinan 250012, China.
| | - Shuna Sun
- Department of Dermatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan 250011, China.
| |
Collapse
|
5
|
Qiu YT, Luo XY, Deng YF, Zheng X, Qiu JG, Zhang LS, Huang XQ, Zheng XB, Huang HY. Modified Pulsatilla decoction alleviates 5-fluorouracil-induced intestinal mucositis by modulating the TLR4/MyD88/NF-κB pathway and gut microbiota. World J Gastroenterol 2025; 31:98806. [PMID: 39991674 PMCID: PMC11755253 DOI: 10.3748/wjg.v31.i7.98806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/19/2024] [Accepted: 12/25/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Modified Pulsatilla decoction (PD), a PD with licorice and ejiao, is a classic Traditional Chinese Medicine formula with significant efficacy in treating intestinal mucositis (IM) induced by tumor therapy. However, its specific molecular and biological mechanisms remain unclear. AIM To investigate the therapeutic effect and mechanism of modified PD in IM. METHODS This study used an IM mouse model established using 5-fluorouracil injections to investigate the effects of the modified PD (3, 6, and 12 g/kg) in IM. The primary chemical components of the modified PD were identified using liquid chromatography-mass spectrometry. Body weight loss, diarrhea scores, intestinal length, histopathological scores, and inflammatory cytokine levels were measured to evaluate the effects of the modified PD in IM. Effects on the TLR4/MyD88/NF-κB pathway were evaluated using western blot analysis. The intestinal microbiota was characterized using Illumina NovaSeq sequencing. RESULTS The results showed that modified PD significantly improved weight loss and diarrhea and shortened the intestines in IM mice. Mechanistically, modified PD suppressed the TLR4/MyD88/NF-κB pathway and downregulated the expression of reactive oxygen species, lipopolysaccharides, and pro-inflammatory cytokines (IL-1β, TNF-α, IFN-γ, IL-6, IL-8, and IL-17), while increasing the expression of the anti-inflammatory cytokine IL-10. Furthermore, modified PD protected the intestinal mucosal barrier by increasing the expression of tight junction proteins (occludin-1, claudin-1, and ZO-1) and mucin-2. Finally, 16S rDNA sequencing revealed that modified PD improved intestinal dysbiosis. CONCLUSION Our research offers new insights into the potential mechanism of modified PD in alleviating IM and provides experimental evidence supporting its pharmaceutical application in clinical IM treatment.
Collapse
Affiliation(s)
- Yi-Tong Qiu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
| | - Xin-Yi Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
- Druggability Research Team, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan 523808, Guangdong Province, China
| | - Ya-Feng Deng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
| | - Xue Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
- Druggability Research Team, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan 523808, Guangdong Province, China
| | - Jian-Guo Qiu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
- Institute of Traditional Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan 523000, Guangdong Province, China
| | - Lin-Sheng Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
| | - Xiao-Qi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
- Druggability Research Team, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan 523808, Guangdong Province, China
| | - Xue-Bao Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 525000, Guangdong Province, China
- Druggability Research Team, Dongguan Institute of Guangzhou University of Chinese Medicine, Dongguan 523808, Guangdong Province, China
| | - Hai-Yang Huang
- Institute of Traditional Chinese Medicine, Dongguan Hospital of Traditional Chinese Medicine, Dongguan 523000, Guangdong Province, China
| |
Collapse
|
6
|
Xie J, Du X, Li Y, Wu C, Li R, Zhao M, Shi S. Berberine shaping the tumor immune landscape via pyroptosis. Cell Immunol 2025; 408:104908. [PMID: 39701005 DOI: 10.1016/j.cellimm.2024.104908] [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: 08/04/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Pyroptosis is a programmed cell death (PCD) mainly mediated by the Gasdermin family of proteins, among which Gasdermin E (GSDME) is considered a tumor suppressor gene. GSDME can recruit immune cells to the tumor microenvironment (TME) and promote their effects. Activating and enhancing adaptive immunity through GSDME is a potential solution for anti-tumor therapy. Here we reported that berberine (BBR), a small molecule from traditional Chinese medicine, as a GSDME activator, induced caspase-3 (C-3)/GSDME pathway-mediated pyroptosis through the mitochondrial pathway, improved the immunosuppressive state of the tumor microenvironment, and thus promoted anti-tumor immunity. We determined the induction of pyroptosis of 4 T1 cells by BBR through various experiments, and investigated the immune activation effect of BBR by co-culture in vitro, which induced DCs maturation and macrophage polarization. Zebrafish embryo toxicity experiments were used to evaluate the in vivo safety of berberine. Furthermore, the in vivo antitumor and immune activation effects of BBR were investigated using 4 T1 orthotopic model mice, and the results showed that BBR could eliminate orthotopic tumor cells by activating local and systemic immunity. Moreover, we observed that BBR significantly inhibited breast cancer lung metastasis. In summary, our results showd the role of BBR as a GSDME activator stimulated both local and systemic antitumor immune responses by inducing pyroptosis, effectively preventing tumor development and metastasis.
Collapse
Affiliation(s)
- Jinjin Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chengyu Wu
- Department of Pharmacy, Shenzhen Technology University, Shenzhen, China.
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Mengnan Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Sanjun Shi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
7
|
Abulsoud AI, Aly SH, Abdel Mageed SS, Abdelmaksoud NM, El-Dakroury WA, Mohammed OA, Abdel-Reheim MA, Zaki MB, Rizk NI, El Tabaa MM, Rashed M, El-Shiekh RA, Doghish AS. Natural compounds as modulators of miRNAs: a new frontier in bladder cancer treatment. Med Oncol 2025; 42:56. [PMID: 39883227 DOI: 10.1007/s12032-025-02613-8] [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: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Bladder cancer (BC) is a major global health issue with a high recurrence rate and limited effective treatments. Over the past few years, it has become evident that miRNAs play a role in the carcinogenesis process, particularly in regulating genes that promote cancer cell proliferation and invasion. This review focuses on the extent to which natural products can act as potential miRNA modulators for the management of bladder cancer. Polyphenols, flavonoids, and other phytochemicals are natural compounds found to have inherent potential to modulate miRNAs and reform the oncogenic properties of bladder cancer cells regulating cell growth and death. In integration with the current cancer treatment regimes, such natural agents may safely substitute for the traditional chemical chemotherapeutic agents of the conventional approaches. To this end, this review presents the existing knowledge of natural compounds as regulators of miRNA, their mechanisms for the management of BC, the role of their nanoparticles, and future novel therapies. The use of these compounds is not only a therapeutic practice for the conditions of bladder cancer, but it also upholds new avenues for creativity.
Collapse
Affiliation(s)
- Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Shaza H Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Nourhan M Abdelmaksoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, 32897, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Menoufia National University, Km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Riham A El-Shiekh
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| |
Collapse
|
8
|
Song Y, Zhao H, Yu R, Zhang Y, Zou Y, Liu X, Sun S. Wogonin suppresses proliferation, invasion and migration in gastric cancer cells via targeting the JAK-STAT3 pathway. Sci Rep 2024; 14:30803. [PMID: 39730467 DOI: 10.1038/s41598-024-81196-2] [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: 05/22/2024] [Accepted: 11/25/2024] [Indexed: 12/29/2024] Open
Abstract
Wogonin is a compound extracted from the medicinal plant Scutellaria baicalensis Geogi and has been found to exert antitumor activities in a variety of malignancies. However, the molecular mechanisms involved in the anti-gastric cancer (GC) effects of wogonin remain poorly understood. In the present study, we found that wogonin treatment inhibited the proliferation of GC cells, induced apoptosis and G0/G1 cell arrest, and suppressed the migration and invasion of SGC-7901 and BGC-823 cells in vitro. In addition, wogonin inhibited in vivo tumor growth in SGC-7901 xenograft mice. Transcriptomic analysis suggested that wogonin affected several signaling pathways closely related to tumor proliferation and metastasis, including the STAT3 signaling pathway. Further research indicated that wogonin may exert antitumor effects in GC cells by downregulating the JAK-STAT3 pathway. Altogether, our results demonstrate that wogonin exerts antitumor effects by perturbing JAK-STAT3 signaling in GC cells and that wogonin may be a potential therapeutic option for GC.
Collapse
Affiliation(s)
- Yang Song
- School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Hui Zhao
- Advanced Medical Research Institute, Shandong University, Jinan, 250012, China
| | - Runze Yu
- The Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Yang Zhang
- Department of Pulmonary and Critial Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology of Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| | - Xiaofei Liu
- Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| | - Shuna Sun
- Department of Dermatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong Provincial Hospital of Traditional Chinese Medicine, The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| |
Collapse
|
9
|
Cao N, Cheng F, Zhou J, Liu N. Identification and construction of prognostic clusters and risk-prognosis model based on aging-immune related genes in bladder cancer. Discov Oncol 2024; 15:742. [PMID: 39630308 PMCID: PMC11618553 DOI: 10.1007/s12672-024-01655-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Faced with the current global ageing situation, advanced age has become a risk factor for bladder carcinogenesis progression and immunotherapy. Exploring the common mechanisms of aging and immune in bladder cancer and finding new prognostic markers and immunotherapeutic targets has become an urgent issue. METHOD Aging-immune related genes (AIGs) were collected from the public databases MSIGDB, HAGR and ImmPort, and hub AIGs were finally identified in the TCGA-BLCA disease cohort by expression, prognosis, and clinicopathological correlation analysis, and the correlation of hub AIGs with immune microenvironment, immunotherapeutic response, ferroptosis and m6A methylation was verified. Subsequently, prognostic clusters and risk-prognosis models for AIGs was constructed by cluster analysis and multifactorial Cox regression analysis, and the gene mutation and immune infiltration characteristics of the different clusters were explored. Finally, the expression level of related genes was verified by immunohistochemical experiments using patient samples from our medical center. RESULT 145 potential prognostic AIGs were collected in bladder cancer and finally clarified NFKB1 and IL7 with significant expression differences, prognostic value and age correlation. By single gene analysis, hub AIGs were explored to be significantly correlated with immunotherapeutic response, immune microenvironment, ferroptosis and m6A methylation. Subsequently, the risk-prognosis model was constructed with Riskscore = (0.0581)*NFKB1 + (- 0.2285)*IL7. And prognostic clusters based on hub AIGs was performed by cluster analysis, which clarified that the high-risk group was the pro-cancer group, which had a lower mutation rate of hub genes and higher of neutrophil infiltration. Finally, immunohistochemistry of patients confirmed that IL7 and NFKB1 were underexpressed in bladder cancer, and the proliferation and migration ability of tumor cells were significantly decreased after overexpression of these genes. CONCLUSION This study is the first to identify NFKB1 and IL7 as hub AIGs in bladder cancer, which provide new prognostic markers and immunotherapeutic targets.
Collapse
Affiliation(s)
- Nihao Cao
- Department of Urology, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - Fei Cheng
- Department of Urology, Nantong Haimen People's Hospital, Nantong, 226100, China
| | - Jincai Zhou
- Department of Urology, Jianhu People's Hospital, Jianhu County, No. 666 South Ring Road, Yancheng, 224700, China.
| | - Ning Liu
- Department of Urology, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao, Hunan Road, Gulou District, Nanjing, 210009, China.
| |
Collapse
|
10
|
Li Y, Xu C, Weng W, Goel A. Combined treatment with Aronia berry extract and oligomeric proanthocyanidins exhibit a synergistic anticancer efficacy through LMNB1-AKT signaling pathways in colorectal cancer. Mol Carcinog 2024; 63:2145-2157. [PMID: 39282961 DOI: 10.1002/mc.23800] [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: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 10/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most prevalent and highly recurrent malignancies worldwide and currently ranks as the second leading cause of cancer-related deaths. The high degree of morbidity and mortality associated with CRC is primarily attributed to the limited effectiveness of current therapeutic approaches and the emergence of chemoresistance to standard treatment modalities. Recent research indicates that several natural products, including Aronia berry extracts (ABE) and oligomeric proanthocyanidins (OPCs), might offer a safe, cost-effective, and multitargeted adjunctive role to cancer treatment. Herein, we hypothesized a combined treatment with ABE and OPCs could synergistically modulate multiple oncogenic pathways in CRC, thereby enhancing their anticancer activity. We initially conducted a series of in vitro experiments to assess the synergistic anticancer effects of ABE and OPCs on CRC cell lines. We demonstrate that these two compounds exhibited a superior synergistic anticancer potential versus individual treatments in enhancing the ability to inhibit cell viability, suppress colony formation, and induce apoptosis (p < 0.05). Consistent with our in vitro findings, we validated this combinatorial anticancer effect in tumor-derived 3D organoids (PDOs; p < 0.01). Using genome-wide transcriptomic profiling, we identified that a specific gene, LMNB1, associated with the cell apoptosis pathway, was found to play a crucial role in exhibiting anticancer effects with these two products. Furthermore, the combined treatment of ABE and OPCs significantly impacted the expression of key proteins involved in apoptosis, including suppressed expression levels of LMNB1 in CRC cell lines (p < 0.05), which resulted in inhibiting downstream AKT phosphorylation. In conclusion, our study provides novel evidence of the synergistic anticancer effects of ABE and OPCs in CRC cells, partially mediated through the regulation of apoptosis and the oncogene LMNB1 within the AKT signaling pathway. These findings have the potential to better appreciate the anticancer potential of natural products in CRC and help improve treatment outcomes in this malignancy.
Collapse
Affiliation(s)
- Yuan Li
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- Department of Clinical Laboratory, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wenhao Weng
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California, USA
- City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
11
|
Abdelhamid AM, Zeinelabdeen Y, Manie T, Khallaf E, Assal RA, Youness RA. miR-17-5p/STAT3/H19: A novel regulatory axis tuning ULBP2 expression in young breast cancer patients. Pathol Res Pract 2024; 263:155638. [PMID: 39388743 DOI: 10.1016/j.prp.2024.155638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND AND AIM UL-16 binding protein 2 (ULBP2) is a highly altered ligand for the activating receptor, NKG2D in breast cancer (BC). However, the mechanism behind its de-regulation in BC patients remains to be explored. The sophisticated crosstalk between miR-17-5p, the lncRNA H19, and STAT3 as a possible upstream regulatory loop for ULBP2 in young BC patients and cell lines remains as an unexplored area. Therefore, this study aimed at unravelling the ncRNA circuit regulating ULBP2 in young BC patients and cell lines. PATIENTS AND METHODS A total of 30 BC patients were recruited for this study. The expression levels of miR-17-5p, lncRNA H19, and STAT3 were examined in 30 BC tissues compared to their normal counterparts. In addition, the expression signatures of those transcripts were compared in young (<40 years) and old BC (≥40 years) patients. miR-17-5p oligonucleotides, STAT3 and H19 siRNAs were transfected in MDA-MB-231 cells using HiPerfect® Transfection Reagent. miR-17-5p and the transcripts of the target genes quantified using RT-qPCR. Their relative expression was calculated using the 2-ΔΔCT method. RESULTS Through acting as a ceRNA circuit that antagonizes the function of miR-17-5p, H19 prevented the miR-17-5p-induced downregulation of STAT3; this mechanism further contributes to the pathogenesis of BC. Ectopic expression of miR-17-5p in MDA-MB-231 cells displayed its prominent role as an indirect potential activator of NK cells by significantly repressing the expression levels of the oncogenic mediator STAT3 and the oncogenic lncRNA H19 and inducing ULBP2 expression level by 3 folds in TNBC cell lines compared to mock cells. Furthermore, knocking down of STAT3 repressed the lncRNA H19 and increased ULBP2 expression levels, whereas siRNAs against H19 increased the expression levels of ULBP2. CONCLUSION This study highlighted the crosstalk between the novel regulatory network composed of miR-17-5p, H19 and STAT3, and their impact on ULBP2 in BC. Moreover, this study underscored the potential role of miR-17-5p in counteracting the immune evasion tactics, particularly the shedding of ULBP2 in young BC patients, through the modulation of the STAT3/H19/ULBP2 regulatory axis. Thus, targeting this novel regulatory network could potentially enhance our understanding and advance the future application of the innate system-mediated immunotherapy in BC.
Collapse
Affiliation(s)
- A M Abdelhamid
- Biotechnology School, Nile University, Giza 12588, Egypt
| | - Y Zeinelabdeen
- Faculty of Medical Sciences/UMCG, University of Groningen, Antonius Deusinglaan 1, Groningen 9713 AV, the Netherlands
| | - T Manie
- Department of Breast Surgery, National Cancer Institute, Cairo University, Cairo, Egypt
| | - E Khallaf
- Department of General Surgery, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - R A Assal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
| | - R A Youness
- Molecular Genetics Research Team (MGRT), Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University, New Administrative Capital 11835, Egypt.
| |
Collapse
|
12
|
Cui Y, Zhou Q, Jin M, Jiang S, Shang P, Dong X, Li L. Research progress on pharmacological effects and bioavailability of berberine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8485-8514. [PMID: 38888754 DOI: 10.1007/s00210-024-03199-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024]
Abstract
Berberine (BBR), a benzylisoquinoline alkaloid obtained from natural medicines such as coptidis rhizoma, has a wide range of pharmacological activities such as protecting the nervous system, protecting the cardiovascular system, anti-inflammatory, antidiabetic, antihyperlipidemic, antitumor, antibacterial, and antidiarrheal. However, factors such as poor solubility, low permeability, P-glycoprotein (P-gp) efflux, and hepatic-intestinal metabolism result in BBR having a low bioavailability (< 1%), which restricts its application in clinical settings. Therefore, improving its bioavailability is a prerequisite for its clinical applications. This review summarizes the various pharmacological effects of BBR and analyzes the main reasons for its poor bioavailability. It introduces methods to improve the bioavailability of BBR through the use of absorption enhancers and P-gp inhibitors, structural modification of BBR, and preparation of BBR salts and cocrystals as well as the development of new formulations and focuses on the bioavailability study of the new formulations of BBR. The research of BBR was also prospected in order to provide reference for the further research of BBR.
Collapse
Affiliation(s)
- Yulong Cui
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Quanying Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Min Jin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siqi Jiang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peizhao Shang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaofan Dong
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lingjun Li
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
13
|
Liu Y, Fang C, Luo J, Gong C, Wang L, Zhu S. Traditional Chinese Medicine for Cancer Treatment. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:583-604. [PMID: 38716616 DOI: 10.1142/s0192415x24500253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In recent years, due to advancements in medical conditions and the development of scientific research, the fundamental research of TCM antitumor treatments has progressed from the cellular level to the molecular and genetic levels. Previous studies have demonstrated the significant role of traditional Chinese medicine (TCM) in antitumor therapy through various mechanisms and pathways. Its mechanism of action is closely associated with cancer biology across different stages. This includes inhibiting tumor cell proliferation, blocking invasion and metastasis to surrounding tissues, inducing tumor cell apoptosis, inhibiting tumor angiogenesis, regulating immune function, maintaining genome stability, preventing mutation, and regulating cell energy metabolism. The use of TCM for eliciting antitumor effects not only has a good therapeutic effect and low side effects, it also provides a solid theoretical basis for clinical treatment and medication. This paper reviews the mechanism of the antitumor effects of TCM based on tumor characteristics. Through our review, we found that TCM not only directly inhibits tumors, but also enhances the body's immunity, thereby indirectly inducing an antitumor effect. This function aligns with the TCM theory of "strengthening the body's resistance to eliminate pathogenic factors". Furthermore, TCM will play a significant role in tumor treatment in clinical settings.
Collapse
Affiliation(s)
- Yangli Liu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Cheng Fang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Jiaojiao Luo
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Chenyuan Gong
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Lixin Wang
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| | - Shiguo Zhu
- Department of Immunology and Pathogenic Biology, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P. R. China
| |
Collapse
|
14
|
Li D, Wang J, Tuo Z, Yoo KH, Yu Q, Miyamoto A, Zhang C, Ye X, Wei W, Wu R, Feng D. Natural products and derivatives in renal, urothelial and testicular cancers: Targeting signaling pathways and therapeutic potential. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 127:155503. [PMID: 38490077 DOI: 10.1016/j.phymed.2024.155503] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Natural products have demonstrated significant potential in cancer drug discovery, particularly in renal cancer (RCa), urothelial carcinoma (UC), and testicular cancer (TC). PURPOSE This review aims to examine the effects of natural products on RCa, UC and TC. STUDY DESIGN systematic review METHODS: PubMed and Web of Science databases were retrieved to search studies about the effects of natural products and derivatives on these cancers. Relevant publications in the reference list of enrolled studies were also checked. RESULTS This review highlighted their diverse impacts on key aspects such as cell growth, apoptosis, metastasis, therapy response, and the immune microenvironment. Natural products not only hold promise for novel drug development but also enhance the efficacy of existing chemotherapy and immunotherapy. Importantly, we exert their effects through modulation of critical pathways and target genes, including the PI3K/AKT pathway, NF-κB pathway, STAT pathway and MAPK pathway, among others in RCa, UC, and TC. CONCLUSION These mechanistic insights provide valuable guidance for researchers, facilitating the selection of promising natural products for cancer management and offering potential avenues for further gene regulation studies in the context of cancer treatment.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea
| | - Qingxin Yu
- Department of pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, 315211, China
| | - Akira Miyamoto
- Department of Rehabilitation, West Kyushu University, Japan
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China
| | - Xing Ye
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, PR China.
| |
Collapse
|
15
|
Khaled AM, Othman MS, Obeidat ST, Aleid GM, Aboelnaga SM, Fehaid A, Hathout HMR, Bakkar AA, Moneim AEA, El-Garawani IM, Morsi DS. Green-Synthesized Silver and Selenium Nanoparticles Using Berberine: A Comparative Assessment of In Vitro Anticancer Potential on Human Hepatocellular Carcinoma Cell Line (HepG2). Cells 2024; 13:287. [PMID: 38334679 PMCID: PMC10854975 DOI: 10.3390/cells13030287] [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: 12/27/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
A well-known natural ingredient found in several medicinal plants, berberine (Ber), has been shown to have anticancer properties against a range of malignancies. The limited solubility and bioavailability of berberine can be addressed using Ber-loaded nanoparticles. In this study, we compared the in vitro cytotoxic effects of both Ber-loaded silver nanoparticles (Ber-AgNPs) and Ber-loaded selenium nanoparticles (Ber-SeNPs) in the human liver cancer cell line (HepG2) and mouse normal liver cells (BNL). The IC50 values in HepG2 for berberine, Ber-AgNPs, Ber-SeNPs, and cisplatin were 26.69, 1.16, 0.04, and 0.33 µg/mL, respectively. Our results show that Ber and its Ag and Se nanoparticles exerted a good antitumor effect against HepG2 cells by inducing apoptosis via upregulating p53, Bax, cytosolic cytochrome C levels, and caspase-3 activity, and the down-regulation of Bcl-2 levels. Similarly, incubation with Ber and both Ber-NPs (Ag and Se) led to a significant dose-dependent elevation in inflammatory markers' (TNF-α, NF-κB, and COX-2) levels compared to the control group. In addition, it led to the arrest of the G1 cell cycle by depleting the expression of cyclin D1 and CDK-2 mRNA. Furthermore, Ber and both Ber-NPs (Ag and Se) caused a significant dose-dependent increase in LDH activity in HepG2 cells. Furthermore, our findings offer evidence that Ber and its nanoparticles intensified oxidative stress in HepG2 cells. Furthermore, the migration rate of cells subjected to berberine and its nanoforms was notably decreased compared to that of control cells. It can be inferred that Ber nanoparticles exhibited superior anticancer efficacy against HepG2 compared to unprocessed Ber, perhaps due to their improved solubility and bioavailability. Furthermore, Ber-SeNPs exhibited greater efficacy than Ber-AgNPs, possibly as a result of the inherent anticancer characteristics of selenium.
Collapse
Affiliation(s)
- Azza M. Khaled
- Biochemistry Department, College of Medicine, University of Ha’il, Hail P.O. Box 2440, Saudi Arabia; (A.M.K.); (M.S.O.); (G.M.A.)
| | - Mohamed S. Othman
- Biochemistry Department, College of Medicine, University of Ha’il, Hail P.O. Box 2440, Saudi Arabia; (A.M.K.); (M.S.O.); (G.M.A.)
| | - Sofian T. Obeidat
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha’il, Hail P.O. Box 2440, Saudi Arabia; (S.T.O.); (S.M.A.)
| | - Ghada M. Aleid
- Biochemistry Department, College of Medicine, University of Ha’il, Hail P.O. Box 2440, Saudi Arabia; (A.M.K.); (M.S.O.); (G.M.A.)
| | - Shimaa M. Aboelnaga
- Basic Sciences Department, Deanship of Preparatory Year, University of Ha’il, Hail P.O. Box 2440, Saudi Arabia; (S.T.O.); (S.M.A.)
| | - Alaa Fehaid
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Mansoura University, El Mansoura 35516, Egypt;
| | - Heba M. R. Hathout
- Natural Resources Department, Faculty of African Postgraduate Studies, Cairo University, Giza 12613, Egypt;
| | - Ashraf A. Bakkar
- Faculty of Biotechnology, October University for Modern Science and Arts (MSA), Giza 12566, Egypt;
| | - Ahmed E. Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Ain Helwan, Cairo 11795, Egypt
| | - Islam M. El-Garawani
- Zoology Department, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt; (I.M.E.-G.); (D.S.M.)
| | - Dalia S. Morsi
- Zoology Department, Faculty of Science, Menoufia University, Shibin El Kom 32511, Egypt; (I.M.E.-G.); (D.S.M.)
| |
Collapse
|
16
|
Ali M, Mishra D, Singh RP. Cancer Pathways Targeted by Berberine: Role of microRNAs. Curr Med Chem 2024; 31:5178-5198. [PMID: 38303534 DOI: 10.2174/0109298673275121231228124031] [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: 07/31/2023] [Revised: 11/26/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024]
Abstract
Cancer is a complex and heterogeneous malignant disease. Due to its multifactorial nature, including progressive changes in genetic, epigenetic, transcript, and protein levels, conventional therapeutics fail to save cancer patients. Evidence indicates that dysregulation of microRNA (miRNA) expression plays a crucial role in tumorigenesis, metastasis, cell proliferation, differentiation, metabolism, and signaling pathways. Moreover, miRNAs can be used as diagnostic and prognostic markers and therapeutic targets in cancer. Berberine, a naturally occurring plant alkaloid, has a wide spectrum of biological activities in different types of cancers. Inhibition of cell proliferation, metastasis, migration, invasion, and angiogenesis, as well as induction of cell cycle arrest and apoptosis in cancer cells, is reported by berberine. Recent studies suggested that berberine regulates many oncogenic and tumor suppressor miRNAs implicated in different phases of cancer. This review discussed how berberine inhibits cancer growth and propagation and regulates miRNAs in cancer cells. And how berberine-mediated miRNA regulation changes the landscape of transcripts and proteins that promote or suppress cancer progression. Overall, the underlying molecular pathways altered by berberine and miRNA influencing the tumor pathophysiology will enhance our understanding to combat the malignancy.
Collapse
Affiliation(s)
- Mansoor Ali
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Deepali Mishra
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rana Pratap Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
17
|
Savoji AB, Kaheni Y, Rezaei P, Farkhondeh T, Pourhanifeh MH, Samarghandian S. Therapeutic Effects of Berberine against Urological Cancers: Biological Potentials Based on Cellular Mechanisms. Curr Mol Med 2024; 24:1282-1290. [PMID: 37933211 DOI: 10.2174/0115665240263630231009050436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Urological cancers, encompassing prostate, kidney, and bladder cancers, pose significant global health challenges. Current treatment modalities, including chemotherapy, radiotherapy, and surgery, individually or in combination, have limitations in efficacy and are associated with notable morbidity and mortality. METHODS This review explores alternative therapeutic avenues, emphasizing the exploration of natural compounds, with a specific focus on berberine. Berberine's potential as a treatment for urological cancers is investigated through an extensive examination of cellular and molecular mechanisms. RESULTS The comprehensive analysis reveals promising anticancer properties associated with berberine, substantiated by a wealth of experimental studies. The agent's impact on urological cancers is discussed, highlighting notable findings related to its efficacy and safety profile. CONCLUSIONS Given the high mortality rates and potential side effects associated with current standard treatments for urological cancers, the exploration of alternative, effective, and safer options is imperative. This review underscores berberine's therapeutic potential, shedding light on its anticancer effects and encouraging further research in the pursuit of enhanced treatment strategies.
Collapse
Affiliation(s)
- Ali Bozorg Savoji
- Student Research Committee, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Yasamin Kaheni
- Student Research Committee, Islamic Azad University, Mashhad Medical Branch, Mashhad, Iran
| | - Pouria Rezaei
- Student Research Committee, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
18
|
Goel A. Current understanding and future prospects on Berberine for anticancer therapy. Chem Biol Drug Des 2023; 102:177-200. [PMID: 36905314 DOI: 10.1111/cbdd.14231] [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: 12/28/2022] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023]
Abstract
Berberine (BBR) is a potential plant metabolite and has remarkable anticancer properties. Many kinds of research are being focused on the cytotoxic activity of berberine in in vitro and in vivo studies. A variety of molecular targets which lead to the anticancer effect of berberine ranges from p-53 activation, Cyclin B expression for arresting cell cycles; protein kinase B (AKT), MAP kinase and IKB kinase for antiproliferative activity; effect on beclin-1 involved in autophagy; reduced expression of MMP-9 and MMP-2 for the inhibition of invasion and metastasis etc. Berberine also interferes with transcription factor-1 (AP-1) activity responsible for the expression of oncogenes and neoplastic transformation of the cell. It also leads to the inhibition of various enzymes which are directly or indirectly involved in carcinogenesis like N acetyl transferase, Cyclo-oxygenase-2, Telomerase and Topoisomerase. In addition to these actions, Berberine plays a role in, the regulation of reactive oxygen species and inflammatory cytokines in preventing cancer formation. Berberine anticancer properties are demonstrated due to the interaction of berberine with micro-RNA. The summarized information presented in this review article may help and lead the researchers, scientists/industry persons to use berberine as a promising candidate against cancer.
Collapse
Affiliation(s)
- Anjana Goel
- Department of Biotechnology, GLA University, Mathura, 281 46, Uttar Pradesh, India
| |
Collapse
|
19
|
Chen Y, Yin L, Hao M, Xu W, Gao J, Sun Y, Wang Q, Chen S, Liang Y, Guo R, Zhang J, Li J, Zhai Q, Cheng R, Wang J, Wang H, Yang Z. Medicarpin induces G1 arrest and mitochondria-mediated intrinsic apoptotic pathway in bladder cancer cells. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:211-225. [PMID: 37307373 DOI: 10.2478/acph-2023-0016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 06/14/2023]
Abstract
Bladder cancer (BC) is the tenth most commonly diagnosed cancer. High recurrence, chemoresistance, and low response rate hinder the effective treatment of BC. Hence, a novel therapeutic strategy in the clinical management of BC is urgently needed. Medicarpin (MED), an isoflavone from Dalbergia odorifera, can promote bone mass gain and kill tumor cells, but its anti-BC effect remains obscure. This study reve aled that MED effectively inhibited the proliferation and arrested the cell cycle at the G1 phase of BC cell lines T24 and EJ-1 in vitro. In addition, MED could significantly suppress the tumor growth of BC cells in vivo. Mechanically, MED induced cell apoptosis by upregulating pro-apoptotic proteins BAK1, Bcl2-L-11, and caspase-3. Our data suggest that MED suppresses BC cell growth in vitro and in vivo via regulating mitochondria-mediated intrinsic apoptotic pathways, which can serve as a promising candidate for BC therapy.
Collapse
Affiliation(s)
- Yuan Chen
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Liqi Yin
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mingxuan Hao
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wenkai Xu
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Jixian Gao
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Yuxin Sun
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Qiao Wang
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Shi Chen
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Youfeng Liang
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
| | - Rui Guo
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jinku Zhang
- 3Department of Pathology, First Central Hospital of Baoding City, Baoding 071000, Hebei, China
- 4Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Baoding 071000, Hebei, China
| | - Jinmei Li
- 3Department of Pathology, First Central Hospital of Baoding City, Baoding 071000, Hebei, China
- 4Key Laboratory of Molecular Pathology and Early Diagnosis of Tumor in Hebei Province, Baoding 071000, Hebei, China
| | - Qiongli Zhai
- 5Department of Pathology, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Runfen Cheng
- 5Department of Pathology, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Jiansong Wang
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Haifeng Wang
- 2Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Zhao Yang
- 1College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing 100029, China
- 6College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar 843300, Xinjiang, China
| |
Collapse
|
20
|
Wu M, Sun J, Wang L, Wang P, Xiao T, Wang S, Liu Q. The lncRNA HOTAIR via miR-17-5p is involved in arsenite-induced hepatic fibrosis through regulation of Th17 cell differentiation. JOURNAL OF HAZARDOUS MATERIALS 2023; 443:130276. [PMID: 36332283 DOI: 10.1016/j.jhazmat.2022.130276] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
Arsenic compounds are toxins that are widely distributed in the environment. Chronic exposure to low levels of these compounds can cause hepatic fibrosis and other damage. Th17 differentiation of CD4+ T cells and the secretion of IL-17 activates hepatic stellate cells (HSCs), which are involved in hepatic fibrosis, but their mechanisms in arsenic-induced hepatic fibrosis are unclear. We found, in arsenite-induced fibrotic livers of mice, increases of CD4+ T cell infiltration, Th17 cell nuclear receptor retinoic acid receptor-related orphan receptor γt (RORγt), and secretion of the pro-inflammatory cytokine IL-17. There were also elevated levels of the lncRNA, HOTAIR. For Jurkat cells, arsenite elevated levels of HOTAIR and protein levels of RORγt and IL-17A, decreased miR-17-5p, promoted Th17 cell differentiation, and released IL-17. The culture medium of arsenite-treated Jurkat cells activated LX-2 cells. Down-regulation of HOTAIR or up-regulation of miR-17-5p blocked arsenite-induced Th17 cell differentiation, which inhibited the LX-2 cell activation. However, down-regulation of HOTAIR and miR-17-5p reversed this inhibitory effect. For mice, silencing of HOTAIR diminished the hepatic levels of RORγt and IL-17A and alleviated arsenite-induced hepatic fibrosis. These results demonstrate that, for CD4+ T cells, arsenite promotes RORγt-mediated Th17 cell differentiation through HOTAIR down-regulation of miR-17-5p, and increases the secretion of cytokine IL-17A, which activates HSCs; the activated HSCs facilitate hepatic fibrosis. The findings reveal a new mechanism and a potential therapeutic target for arsenite-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Department of Nutrition, Functional Food Clinical Evaluation Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, Jiangsu, People's Republic of China
| | - Li Wang
- Department of Toxicology, School of Public Health, Baotou Medical College, Baotou 014040, Inner Mongolia, People's Republic of China
| | - Peiwen Wang
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Tian Xiao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Suhua Wang
- Department of Toxicology, School of Public Health, Baotou Medical College, Baotou 014040, Inner Mongolia, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Suzhou Institute of Public Health, Gusu School, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Medicine, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
21
|
El-Mahdy HA, Elsakka EGE, El-Husseiny AA, Ismail A, Yehia AM, Abdelmaksoud NM, Elshimy RAA, Noshy M, Doghish AS. miRNAs role in bladder cancer pathogenesis and targeted therapy: Signaling pathways interplay - A review. Pathol Res Pract 2023; 242:154316. [PMID: 36682282 DOI: 10.1016/j.prp.2023.154316] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Bladder cancer (BC) is the 11th most popular cancer in females and 4th in males. A lot of efforts have been exerted to improve BC patients' care. Besides, new approaches have been developed to enhance the efficiency of BC diagnosis, prognosis, therapeutics, and monitoring. MicroRNAs (miRNAs, miRs) are small chain nucleic acids that can regulate wide networks of cellular events. They can inhibit or degrade their target protein-encoding genes. The miRNAs are either downregulated or upregulated in BC due to epigenetic alterations or biogenesis machinery abnormalities. In BC, dysregulation of miRNAs is associated with cell cycle arrest, apoptosis, proliferation, metastasis, treatment resistance, and other activities. A variety of miRNAs have been related to tumor kind, stage, or patient survival. Besides, although new approaches for using miRNAs in the diagnosis, prognosis, and treatment of BC have been developed, it still needs further investigations. In the next words, we illustrate the recent advances in the role of miRNAs in BC aspects. They include the role of miRNAs in BC pathogenesis and therapy. Besides, the clinical applications of miRNAs in BC diagnosis, prognosis, and treatment are also discussed.
Collapse
Affiliation(s)
- Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Amr Mohamed Yehia
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Nourhan M Abdelmaksoud
- Department of Biochemistry and Biotechnology, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Reham A A Elshimy
- Clinical & Chemical Pathology Department, National Cancer Institute, Cairo University, 11796 Cairo, Egypt
| | - Mina Noshy
- Clinical Pharmacy Department, Faculty of Pharmacy, King Salman International University (KSIU), SouthSinai, Ras Sudr 46612, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| |
Collapse
|
22
|
Bernitsa S, Dayan R, Stephanou A, Tzvetanova ID, Patrikios IS. Natural biomolecules and derivatives as anticancer immunomodulatory agents. Front Immunol 2023; 13:1070367. [PMID: 36700235 PMCID: PMC9868674 DOI: 10.3389/fimmu.2022.1070367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Despite advancements in chemotherapy, the issue of resistance and non-responsiveness to many chemotherapeutic drugs that are currently in clinical use still remains. Recently, cancer immunotherapy has gathered attention as a novel treatment against select cancers. Immunomodulation is also emerging as an effective strategy to improve efficacy. Natural phytochemicals, with known anticancer properties, been reported to mediate their effects by modulating both traditional cancer pathways and immunity. The mechanism of phytochemical mediated-immunomodulatory activity may be attributed to the remodeling of the tumor immunosuppressive microenvironment and the sensitization of the immune system. This allows for improved recognition and targeting of cancer cells by the immune system and synergy with chemotherapeutics. In this review, we will discuss several well-known plant-derived biomolecules and examine their potential as immunomodulators, and therefore, as novel immunotherapies for cancer treatment.
Collapse
Affiliation(s)
| | - Rotem Dayan
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | | | | | | |
Collapse
|
23
|
Zhu C, Li K, Peng XX, Yao TJ, Wang ZY, Hu P, Cai D, Liu HY. Berberine a traditional Chinese drug repurposing: Its actions in inflammation-associated ulcerative colitis and cancer therapy. Front Immunol 2022; 13:1083788. [PMID: 36561763 PMCID: PMC9763584 DOI: 10.3389/fimmu.2022.1083788] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Berberine (BBR), an isoquinoline alkaloid extracted from Coptidis Rhizoma, has a long history of treating dysentery in the clinic. Over the past two decades, the polytrophic, pharmacological, and biochemical properties of BBR have been intensively studied. The key functions of BBR, including anti-inflammation, antibacterial, antioxidant, anti-obesity, and even antitumor, have been discovered. However, the underlying mechanisms of BBR-mediated regulation still need to be explored. Given that BBR is also a natural nutrition supplement, the modulatory effects of BBR on nutritional immune responses have attracted more attention from investigators. In this mini-review, we summarized the latest achievements of BBR on inflammation, gut microbes, macrophage polarization, and immune responses associated with their possible tools in the pathogenesis and therapy of ulcerative colitis and cancer in recent 5 years. We also discuss the therapeutic efficacy and anti-inflammatory actions of BBR to benefit future clinical applications.
Collapse
Affiliation(s)
- Cuipeng Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Kaiqi Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xiao-Xu Peng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tong-Jia Yao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zi-Yu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China,*Correspondence: Hao-Yu Liu, ; Demin Cai,
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China,Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China,*Correspondence: Hao-Yu Liu, ; Demin Cai,
| |
Collapse
|
24
|
Duan B, Yu Z, Liu R, Li J, Song Z, Zhou Q, Chen L. Tetrandrine-induced downregulation of lncRNA NEAT1 inhibits rheumatoid arthritis progression through the STAT3/miR-17-5p pathway. Immunopharmacol Immunotoxicol 2022; 44:886-893. [PMID: 35815670 DOI: 10.1080/08923973.2022.2092748] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The inhibitory effect of Tetrandrine (Tet) on rheumatoid arthritis (RA) is well established. However, its exact molecular mechanism remains unknown. METHODS RT-qPCR coupled with western blotting was employed to analyze the expression of NEAT1, miR-17-5p, and STAT3 in RA tissues and/or RA-fibroblast-like synoviocytes (RA-FLS) treated with 3 μmol/L of Tet for 48 h. Cell Counting Kit-8 assay and flow cytometry were performed to assess RA-FLS proliferation and apoptosis. Luciferase reporter assays were used to validate the interactions between miR-17-5p and STAT3 or NEAT1. RESULTS The expression of NEAT1 decreased in a time-dependent manner upon Tet treatment. Tet significantly inhibited RA-FLS proliferation and triggered apoptosis by downregulating NEAT1 expression. Additionally, NEAT1 directly targeted miR-17-5p to upregulate STAT3 expression. Tet-induced low NEAT1 expression impaired RA-FLS growth by targeting miR-17-5p and inhibiting STAT3. CONCLUSION Tet exerts its inhibitory role in RA progression by regulating the NEAT1/miR-17-5p/STAT3 pathway.
Collapse
Affiliation(s)
- Bo Duan
- Department of Rheumatology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Zhao Yu
- Department of Rheumatology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Ruilin Liu
- Department of Rheumatology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jigao Li
- Department of Rheumatology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhe Song
- Department of Rheumatology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Quan Zhou
- Department of Rheumatology, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Lichuan Chen
- Department of Rheumatology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
25
|
Crosstalk of miRNAs with signaling networks in bladder cancer progression: Therapeutic, diagnostic and prognostic functions. Pharmacol Res 2022; 185:106475. [DOI: 10.1016/j.phrs.2022.106475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/27/2022] [Indexed: 12/24/2022]
|
26
|
Berberine: An Important Emphasis on Its Anticancer Effects through Modulation of Various Cell Signaling Pathways. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185889. [PMID: 36144625 PMCID: PMC9505063 DOI: 10.3390/molecules27185889] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 11/27/2022]
Abstract
Cancer is the most commonly diagnosed type of disease and a major cause of death worldwide. Despite advancement in various treatment modules, there has been little improvement in survival rates and side effects associated with this disease. Medicinal plants or their bioactive compounds have been extensively studied for their anticancer potential. Novel drugs based on natural products are urgently needed to manage cancer through attenuation of different cell signaling pathways. In this regard, berberine is a bioactive alkaloid that is found in variety of plants, and an inverse association has been revealed between its consumption and cancer. Berberine exhibits an anticancer role through scavenging free radicals, induction of apoptosis, cell cycle arrest, inhibition of angiogenesis, inflammation, PI3K/AKT/mammalian target of rapamycin (mTOR), Wnt/β-catenin, and the MAPK/ERK signaling pathway. In addition, synergistic effects of berberine with anticancer drugs or natural compounds have been proven in several cancers. This review outlines the anticancer effects and mechanisms of action of berberine in different cancers through modulation of various cell signaling pathways. Moreover, the recent developments in the drug delivery systems and synergistic effect of berberine are explained.
Collapse
|
27
|
Xiong RG, Huang SY, Wu SX, Zhou DD, Yang ZJ, Saimaiti A, Zhao CN, Shang A, Zhang YJ, Gan RY, Li HB. Anticancer Effects and Mechanisms of Berberine from Medicinal Herbs: An Update Review. Molecules 2022; 27:4523. [PMID: 35889396 PMCID: PMC9316001 DOI: 10.3390/molecules27144523] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/25/2022] Open
Abstract
Cancer has been a serious public health problem. Berberine is a famous natural compound from medicinal herbs and shows many bioactivities, such as antioxidant, anti-inflammatory, antidiabetic, anti-obesity, and antimicrobial activities. In addition, berberine shows anticancer effects on a variety of cancers, such as breast, lung, gastric, liver, colorectal, ovarian, cervical, and prostate cancers. The underlying mechanisms of action include inhibiting cancer cell proliferation, suppressing metastasis, inducing apoptosis, activating autophagy, regulating gut microbiota, and improving the effects of anticancer drugs. This paper summarizes effectiveness and mechanisms of berberine on different cancers and highlights the mechanisms of action. In addition, the nanotechnologies to improve bioavailability of berberine are included. Moreover, the side effects of berberine are also discussed. This paper is helpful for the prevention and treatment of cancers using berberine.
Collapse
Affiliation(s)
- Ruo-Gu Xiong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Si-Yu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Si-Xia Wu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Dan-Dan Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Zhi-Jun Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Adila Saimaiti
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| | - Cai-Ning Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China;
| | - Ao Shang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China;
| | - Yun-Jian Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Ren-You Gan
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, National Agricultural Science & Technology Center, Chengdu 610213, China;
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (R.-G.X.); (S.-Y.H.); (S.-X.W.); (D.-D.Z.); (Z.-J.Y.); (A.S.)
| |
Collapse
|
28
|
Sun JX, Liu CQ, Xu JZ, An Y, Xu MY, Zhong XY, Zeng N, Ma SY, He HD, Zhang ZB, Wang SG, Xia QD. A Four-Cell-Senescence-Regulator-Gene Prognostic Index Verified by Genome-Wide CRISPR Can Depict the Tumor Microenvironment and Guide Clinical Treatment of Bladder Cancer. Front Immunol 2022; 13:908068. [PMID: 35898492 PMCID: PMC9312376 DOI: 10.3389/fimmu.2022.908068] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/08/2022] [Indexed: 01/10/2023] Open
Abstract
Bladder cancer (BCa) is the 10th most commonly diagnosed cancer worldwide, and cellular senescence is defined as a state of permanent cell cycle arrest and considered to play important roles in the development and progression of tumor. However, the comprehensive effect of senescence in BCa has not ever been systematically evaluated. Using the genome-wide CRISPR screening data acquired from DepMap (Cancer Dependency Map), senescence genes from the CellAge database, and gene expression data from The Cancer Genome Atlas (TCGA), we screened out 12 senescence genes which might play critical roles in BCa. A four-cell-senescence-regulator-gene prognostic index was constructed using the least absolute shrinkage and selection operator (LASSO) and multivariate COX regression model. The transcriptomic data and clinical information of BCa patients were downloaded from TCGA and Gene Expression Omnibus (GEO). We randomly divided the patients in TCGA cohort into training and testing cohorts and calculated the risk score according to the expression of the four senescence genes. The validity of this risk score was validated in the testing cohort (TCGA) and validation cohort (GSE13507). The Kaplan–Meier curves revealed a significant difference in the survival outcome between the high- and low-risk score groups. A nomogram including the risk score and other clinical factors (age, gender, stage, and grade) was established with better predictive capacity of OS in 1, 3, and 5 years. Besides, we found that patients in the high-risk group had higher tumor mutation burden (TMB); lower immune, stroma, and ESTIMATE scores; higher tumor purity; aberrant immune functions; and lower expression of immune checkpoints. We also performed gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA) to investigate the interaction between risk score and hallmark pathways and found that a high risk score was connected with activation of senescence-related pathways. Furthermore, we found that a high risk score was related to better response to immunotherapy and chemotherapy. In conclusion, we identified a four-cell-senescence-regulator-gene prognostic index in BCa and investigated its relationship with TMB, the immune landscape of tumor microenvironment (TME), and response to immunotherapy and chemotherapy, and we also established a nomogram to predict the prognosis of patients with BCa.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zong-Biao Zhang
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| | - Shao-Gang Wang
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| | - Qi-Dong Xia
- *Correspondence: Zong-Biao Zhang, ; Shao-Gang Wang, ; Qi-Dong Xia,
| |
Collapse
|
29
|
Wu W, Lu P, Huang Y, Zhu Z, Li C, Liu Y. Emodin regulates the autophagy via the miR-371a-5p/PTEN axis to inhibit hepatic malignancy. Biochem Biophys Res Commun 2022; 619:1-8. [PMID: 35724456 DOI: 10.1016/j.bbrc.2022.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/27/2022] [Accepted: 06/03/2022] [Indexed: 11/28/2022]
Abstract
Emodin has been reported to fulfill an important function in suppressing the vicious outcome of liver cancer. We aimed to elucidate the partial underlying molecular mechanism of emodin in inhibiting liver cancer, and we applied miRNA-sequence analysis and corresponding molecular functional experiments to find that the inhibitory effect of emodin on liver cancer was partly mediated by cellular autophagy through the miR-371a-5p/PTEN axis. The expression level of miR-371a-5p was down-regulated after emodin treatment in liver cancer cell lines (LCCLs). Restoring the expression level of miR-371a-5p attenuated the suppression of emodin on LCCLs. Additionally, we performed the prediction in relevant online databases and found that PTEN might functioned as a downstream target of miR-371a-5p to participate in the regulation on the above process. What's more, the detection of autophagy-related protein markers showed that LC3II was elevated accompanied by the decreased P62. The above results revealed that PTEN functioned as a key target to regulate the autophagy in the process where emodin inhibited the malignant outcome of LCCLs via miR-371a-5p, which further provided a theoretical basis for the application of traditional Chinese medicine (TCM) on clinical tumors.
Collapse
Affiliation(s)
- Wu Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peilin Lu
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yujing Huang
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunming Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Liu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Du Y, Khan M, Fang N, Ma F, Du H, Tan Z, Wang H, Yin S, Wei X. Berberine Attenuates Cell Motility via Inhibiting Inflammation-Mediated Lysyl Hydroxylase-2 and Glycolysis. Front Pharmacol 2022; 13:856777. [PMID: 35559258 PMCID: PMC9086160 DOI: 10.3389/fphar.2022.856777] [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: 01/17/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Lysyl hydroxylase-2 (LH2) involves in the hydroxylation of telopeptide lysine residues during collagen deposition. Recent studies indicate that interleukin (IL)-6 generated by the chronic inflammation disease may trigger the LH2 expression to accelerate cell motility. Berberine is the alkaloid derived from the traditional Chinese medicine Coptis chinensis, which displays potential anti-inflammatory activity in multiple diseases. The anti-inflammatory activity of berberine has been confirmed by reducing proinflammatory cytokines such as IL-6, IL-8, and IFN-γ. However, whether and how berberine inhibits cellular motility against metastatic spread in triple-negative breast cancer (TNBC) has not been demonstrated, and the underlying mechanism remains unclear. We investigated the effects of berberine on the inflammatory cytokine secretion, cell proliferation, and migration in vitro and further explored the effect of berberine on growth and metastasis in vivo. Berberine restrained TNBC cell proliferation, motility, and glycolysis process in a dose-dependent way. The secretion of IL-6 was abrogated by berberine in TNBC cells, and IL-6-stimulated cell migration was inhibited by berberine. Mechanistically, berberine remarkably suppressed LH2 expression at both mRNA and protein levels. LH2 depletion led to decreasing the antimotility effect of berberine, and this phenomenon was related to the suppressed glycolysis after LH2 inhibition. Conversely, ectopic restoration of LH2 could further increase the antimotility effect of berberine. Moreover, berberine was confirmed to inhibit cell growth and motility in vivo, and the expression of LH2 and glycolytic enzymes was also blocked by berberine in vivo. Collectively, this study indicated that berberine could be a promising therapeutic drug via regulating LH2 for TNBC.
Collapse
Affiliation(s)
- Yishan Du
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Muhammad Khan
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Nana Fang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Hefei, China
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhenya Tan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shi Yin
- Department of Geriatrics, Affiliated Provincial Hospital of Anhui Medical University, Anhui Medical University, Hefei, China.,Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei, China
| | - Xiaohui Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
31
|
Zhao X, Cui D, Yuan W, Chen C, Liu Q. Berberine represses Wnt/β-catenin pathway activation via modulating the microRNA-103a-3p/Bromodomain-containing protein 4 axis, thereby refraining pyroptosis and reducing the intestinal mucosal barrier defect induced via colitis. Bioengineered 2022; 13:7392-7409. [PMID: 35259053 PMCID: PMC8973728 DOI: 10.1080/21655979.2022.2047405] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Intestinal barrier dysfunction is inflammatory bowel disease’s hallmark. Berberine (BBR) has manifested its anti-inflammatory properties in colitis. For exploring the molecular mechanism of BBR’s impacts on colitis, application of a dextran sodium sulfate-induced mouse colitis in vivo model was with recording the body weight, stool consistency, stool occult blood and general physical symptoms of all groups of mice every day. Behind assessment of intestinal permeability, detection of colon damage’s degree and apoptosis, and inflammatory factors for assessment of pyroptosis was conducted. Application of interleukin-6-stimulated Caco-2 cells was for construction of an in vitro model. Then detection of cell advancement with inflammation and measurement of the barrier’s integrity were put into effect. Verification of microRNA (miR)-103a-3p and Bromodomain-containing protein 4 (BRD4)’s targeting link was conducted. Experiments have clarified BBR, elevated miR-103a-3p or repressive BRD4 was available to alleviate colitis-stimulated pyroptosis and intestinal mucosal barrier defects. BBR elevated miR-103a-3p to target BRD4; Refraining miR-103a-3p or enhancive BRD4 turned around BBR’s therapeutic action on colitis injury. BBR depressed Wnt/β-catenin pathway activation via controlling the miR-103a-3p/BRD4 axis. All in all, BBR represses Wnt/β-catenin pathway activation via modulating the miR-103a-3p/BRD4 axis, thereby mitigating colitis-stimulated pyroptosis and the intestinal mucosal barrier defect. The research suggests BBR is supposed to take on potential in colitis cure.
Collapse
Affiliation(s)
- Xun Zhao
- The Graduate School, Guizhou Medical University, Guiyang City, Guizhou Province, China
| | - DeJun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang City, Guizhou Province, China
| | - WenQiang Yuan
- The Graduate School, Guizhou Medical University, Guiyang City, Guizhou Province, China
| | - Chen Chen
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang City, Guizhou Province, China
| | - Qi Liu
- The Graduate School, Guizhou Medical University, Guiyang City, Guizhou Province, China
| |
Collapse
|
32
|
Han W, Kong D, Lu Q, Zhang W, Fan Z. Aloperine inhibits colorectal cancer cell proliferation and metastasis progress via regulating miR-296-5p/STAT3 axis. Tissue Cell 2021; 74:101706. [PMID: 34883316 DOI: 10.1016/j.tice.2021.101706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 11/18/2021] [Accepted: 11/27/2021] [Indexed: 11/26/2022]
Abstract
Anti-tumorous effect of Aloperine (ALO) has been previously found. This study examined the role and the underlying mechanism of ALO in colorectal cancer (CRC). CRC cells were processed by different concentrations of ALO, and subsequently the cell proliferation was detected by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and miR-296-5p expression was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Moreover, the target gene of miR-296-5p was predicted by TargetScan and confirmed by dual-luciferase reporter assay. The expressions of signal transducer and activator of transcription 3 (STAT3), apoptosis-related proteins and epithelial-mesenchymal transition (EMT)-related markers were measured by Western blot. Clone formation assay, flow cytometry, wound-healing and Transwell assays were respectively employed to detect cell proliferation, apoptosis, migration and invasion. ALO inhibited CRC cell proliferation in a dose-dependent manner. MiR-296-5p was low-expressed in CRC tissues and cells, and ALO promoted miR-296-5p expression. STAT3 was targeted by miR-296-5p. Up-regulation of miR-296-5p and ALO treatment both suppressed STAT3 expression, inhibited CRC cell proliferation, migration, invasion as well as the expressions of Bcl-2 and N-cadherin, but promoted apoptosis and expressions of Bax and E-cadherin, which were all reversed by overexpressed STAT3. ALO inhibited CRC cell proliferation, metastasis and EMT but promoted apoptosis via regulating miR-296-5p/STAT3 axis.
Collapse
Affiliation(s)
- Wei Han
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China; Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China; Department of General Surgery, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China
| | - Qin Lu
- Department of Proctology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China
| | - Wei Zhang
- Department of Anesthesiology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China
| | - Zhimin Fan
- Department of Proctology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210012, China.
| |
Collapse
|