1
|
Chen Z, Zheng X, Zeng W, Wang J, Lin Q. JAK2 inactivating mutations promotes endometrial cancer progression by targeting HIF-1α. Discov Oncol 2024; 15:836. [PMID: 39720955 DOI: 10.1007/s12672-024-01722-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024] Open
Abstract
OBJECTIVE Endometrial cancer (EC) is the ninth most common malignancy among women. While mutations in JAK2 are frequently observed in EC, the specific biological functions of JAK2 in endometrial cancer are poorly understood. METHODS The genetic alterations of JAK2 in different cancer types were explored using sequencing dataset deposited at TCGA database. JAK2 mutations were detected in EC formalin-fixed paraffin-embedded (FFPE) samples using Sanger sequencing. The expression levels of JAK2 was accessed using the TCGA database and immunohistochemistry. Furthermore, the relationships between JAK2 expression and staging and prognosis of EC patients were investigated using the TCGA database. Down-regulation of JAK2 were achieved by transient transfection with short hairpin RNAs (shRNAs). Effects of JAK2 on cancer cells proliferation and migration were evaluated by CCK8, colony formation, and transwell assay. The potential biological functions of JAK2 in EC were identified based on bioinformatics analysis. Effects of JAK2 on expression levels of target genes were detected by RT-qPCR and western blotting. Co-immunoprecipitation (co-IP) assays was used to detect the physical association between JAK2 and HIF-1α. RESULTS Frequent mutations and down-regulation of JAK2 were found in EC. Loss-of-function (LOF) assays suggested that JAK2 silencing in endometrial cancer cells promoted cell proliferation and migration, which were partially dependent on HIF-1α signaling pathway. Furthermore, our findings demonstrated that JAK2 interacted with HIF-1α and reduced HIF1α protein expression under hypoxia. CONCLUSION These findings revealed novel molecular mechanisms underlying JAK2 LOF mutations-driven endometrial tumorigenesis and revealed that the HIF-1α pathway may be a potential therapeutic target in JAK2-mutated endometrial cancer.
Collapse
Affiliation(s)
- Zheng Chen
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
| | - Xuan Zheng
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
| | - Weijian Zeng
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
- Shanghai Municipal Key Clinical Specialty, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China
| | - Juan Wang
- Hangzhou Chexmed Technology Co., Ltd., Hangzhou, 310000, China
| | - Qin Lin
- Department of Obstetrics and Gynecology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China.
- Shanghai Municipal Key Clinical Specialty, The International Peace Maternity and Child Health Hospital, Shanghai, 200030, China.
| |
Collapse
|
2
|
Su Y, Liu L, Lin C, Deng D, Li Y, Huang M, Wang Y, Ling K, Wang H, Chen Q, Huang G. Enhancing cancer therapy: advanced nanovehicle delivery systems for oridonin. Front Pharmacol 2024; 15:1476739. [PMID: 39691396 PMCID: PMC11649421 DOI: 10.3389/fphar.2024.1476739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Oridonin (ORI), an ent-kaurane diterpenoid derived from Rabdosia rubescens (Hemsl.) H.Hara, serves as the primary bioactive component of this plant. It demonstrates a broad spectrum of therapeutic activities, including moderate to potent anticancer properties, alongside anti-inflammatory, antibacterial, antifibrotic, immunomodulatory, and neuromodulatory effects, thus influencing diverse biological processes. However, its clinical potential is significantly constrained by poor aqueous solubility and limited bioavailability. In alignment with the approach of developing drug candidates from natural compounds, various strategies, such as structural modification and nanocarrier systems, have been employed to address these challenges. This review provides an overview of ORI-based nano-delivery systems, emphasizing their potential to improve the clinical applicability of oridonin in oncology. Although some progress has been made in advancing ORI nano-delivery research, it remains insufficient for clinical implementation, necessitating further investigation.
Collapse
Affiliation(s)
- Yilin Su
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Lisha Liu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Chongyang Lin
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Dashi Deng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yunfei Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Mou Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yu Wang
- Institute of Pain, The Affiliated Hospital of Southwest Jiaotong University, The Chengdu Third People’s Hospital, Chengdu, China
| | - Kangqiu Ling
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Haobing Wang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Qiyu Chen
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Guixiao Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Zhou M, Zhang Y, Zhang Q, Tong Y. METTL14-mediated m6A modification upregulated SOCS3 expression alleviates thyroid cancer progression by regulating the JAK2/STAT3 pathway. Mol Cell Probes 2024; 78:101987. [PMID: 39413935 DOI: 10.1016/j.mcp.2024.101987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/14/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Thyroid cancer (TC) is the most common malignant tumor of the head and neck. As a common epigenetic modification in mRNAs, N6-methyladenosine (m6A) modification plays critical roles in biological process of cancers. However, m6A methyltransferase methyltransferase-like 14 (METTL14)-mediated m6A modification and its potential regulatory mechanisms in TC are not fully elucidated. In our study, we observed that METTL14 was decreased in TC tissues and cells. And upregulation of METTL14 induced apoptotic cell death and hampered cell proliferation, epithelial mesenchymal transition (EMT) and tumor growth in vitro and in vivo. Mechanistically, METTL14 increased the expression of suppressor of cytokine signaling 3 (SOCS3) through m6A methylation modification, and knockdown of SOCS3 reversed the inhibitory effect of overexpressing METTL14 on TC tumorigenesis. In addition, METTL14-mediated m6A modification of SOCS3 inactivated the janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) pathway, and in the METTL14-overexpressing TC cells, silencing SOCS3-induced upregulation of cell proliferation, EMT and suppression of apoptosis was reversed by JAK2/STAT3 inhibitor AG490 and WP1066. Together, we indicated that METTL14/m6A/SOCS3/JAK2/STAT3 axis play an important role in the progression of TC.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Thyroid Vascular Surgery, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, China.
| | - Yaqi Zhang
- Department of Oncology, Huanggang Central Hospital, Huanggang, 438000, China.
| | - Qiong Zhang
- Department of Dermatology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, China.
| | - Yanchu Tong
- Department of Thyroid Vascular Surgery, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434000, China.
| |
Collapse
|
4
|
Hua X, Xu Q, Wu R, Sun W, Gu Y, Zhu S, Liu X, Lv T, Song Y. ALKBH5 promotes non-small cell lung cancer progression and susceptibility to anti-PD-L1 therapy by modulating interactions between tumor and macrophages. J Exp Clin Cancer Res 2024; 43:164. [PMID: 38872221 PMCID: PMC11177518 DOI: 10.1186/s13046-024-03073-0] [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: 02/23/2024] [Accepted: 05/19/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Understanding the mechanisms that mediate the interaction between tumor and immune cells may provide therapeutic benefit to patients with cancer. The N6-methyladenosine (m6A) demethylase, ALKBH5 (alkB homolog 5), is overexpressed in non-small cell lung cancer. However, its role in the tumor microenvironment is unknown. METHODS Datasets and tissue samples were used to determine the relationship between ALKBH5 expression and immunotherapy efficacy. Bioinformatic analysis, colorimetric assay to determine m6A RNA methylation, dual luciferase reporter assay, RNA/m6A-modified RNA immunoprecipitation, RNA stability assay, and RNA sequencing were used to investigate the regulatory mechanism of ALKBH5 in non-small cell lung cancer. In vitro and in vivo assays were performed to determine the contribution of ALKBH5 to the development of non-small cell lung cancer. RESULTS ALKBH5 was upregulated in primary non-small cell lung cancer tissues. ALKBH5 was positively correlated with programmed death-ligand 1 expression and macrophage infiltration and was associated with immunotherapy response. JAK2 was identified as a target of ALKBH5-mediated m6A modification, which activates the JAK2/p-STAT3 pathway to promote non-small cell lung cancer progression. ALKBH5 was found to recruit programmed death-ligand 1-positive tumor-associated macrophages and promote M2 macrophage polarization by inducing the secretion of CCL2 and CXCL10. ALKBH5 and tumor-associated macrophage-secreted IL-6 showed a synergistic effect to activate the JAK2/p-STAT3 pathway in cancer cells. CONCLUSIONS ALKBH5 promotes non-small cell lung cancer progression by regulating cancer and tumor-associated macrophage behavior through the JAK2/p-STAT3 pathway and the expression of CCL2 and CXCL10, respectively. These findings suggest that targeting ALKBH5 is a promising strategy of enhancing the anti-tumor immune response in patients with NSCLC and that identifying ALKBH5 status could facilitate prediction of clinical response to anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Xin Hua
- Medical School of Southeast University, Nanjing, 210003, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Qiuli Xu
- Medical School of Southeast University, Nanjing, 210003, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Ranpu Wu
- Medical School of Southeast University, Nanjing, 210003, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Wei Sun
- Medical School of Southeast University, Nanjing, 210003, China
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Yanli Gu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Xin Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China
| | - Tangfeng Lv
- Medical School of Southeast University, Nanjing, 210003, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Yong Song
- Medical School of Southeast University, Nanjing, 210003, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
5
|
Di Dalmazi G, Giuliani C, Bucci I, Mascitti M, Napolitano G. Promising Role of Alkaloids in the Prevention and Treatment of Thyroid Cancer and Autoimmune Thyroid Disease: A Comprehensive Review of the Current Evidence. Int J Mol Sci 2024; 25:5395. [PMID: 38791433 PMCID: PMC11121374 DOI: 10.3390/ijms25105395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Thyroid cancer (TC) and thyroid autoimmune disorders (AITD) are among the most common diseases in the general population, with higher incidence in women. Chronic inflammation and autoimmunity play a pivotal role in carcinogenesis. Some studies, indeed, have pointed out the presence of AITD as a risk factor for TC, although this issue remains controversial. Prevention of autoimmune disease and cancer is the ultimate goal for clinicians and scientists, but it is not always feasible. Thus, new treatments, that overcome the current barriers to prevention and treatment of TC and AITD are needed. Alkaloids are secondary plant metabolites endowed with several biological activities including anticancer and immunomodulatory properties. In this perspective, alkaloids may represent a promising source of prophylactic and therapeutic agents for TC and AITD. This review encompasses the current published literature on alkaloids effects on TC and AITD, with a specific focus on the pathways involved in TC and AITD development and progression.
Collapse
Affiliation(s)
- Giulia Di Dalmazi
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Cesidio Giuliani
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Ines Bucci
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Mascitti
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgio Napolitano
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (C.G.); (I.B.); (G.N.)
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
6
|
Ali MA, Khan N, Ali A, Akram H, Zafar N, Imran K, Khan T, Khan K, Armaghan M, Palma‐Morales M, Rodríguez‐Pérez C, Caunii A, Butnariu M, Habtemariam S, Sharifi‐Rad J. Oridonin from Rabdosia rubescens: An emerging potential in cancer therapy - A comprehensive review. Food Sci Nutr 2024; 12:3046-3067. [PMID: 38726411 PMCID: PMC11077219 DOI: 10.1002/fsn3.3986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 05/12/2024] Open
Abstract
Cancer incidences are rising each year. In 2020, approximately 20 million new cancer cases and 10 million cancer-related deaths were recorded. The World Health Organization (WHO) predicts that by 2024 the incidence of cancer will increase to 30.2 million individuals annually. Considering the invasive characteristics of its diagnostic procedures and therapeutic methods side effects, scientists are searching for different solutions, including using plant-derived bioactive compounds, that could reduce the probability of cancer occurrence and make its treatment more comfortable. In this regard, oridonin (ORI), an ent-kaurane diterpenoid, naturally found in the leaves of Rabdosia rubescens species, has been found to have antitumor, antiangiogenesis, antiasthmatic, antiinflammatory, and apoptosis induction properties. Extensive research has been performed on ORI to find various mechanisms involved in its anticancer activities. This review article provides an overview of ORI's effectiveness on murine and human cancer populations from 1976 to 2022 and provides insight into the future application of ORI in different cancer therapies.
Collapse
Affiliation(s)
| | - Noohela Khan
- Department of Nutrition SciencesRashid Latif Medical CollegeLahorePakistan
| | - Ahmad Ali
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Hira Akram
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Noushaba Zafar
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Kinza Imran
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Tooba Khan
- Department of Healthcare Biotechnology, Atta‐ur‐Rahman School of Applied BiosciencesNational University of Sciences and TechnologyIslamabadPakistan
| | | | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta‐ur‐Rahman School of Applied BiosciencesNational University of Sciences and TechnologyIslamabadPakistan
| | - Marta Palma‐Morales
- Departamento de Nutrición y Bromatología, Facultad de FarmaciaUniversidad de GranadaGranadaSpain
- Instituto de Nutrición y Tecnología de los Alimentos ‘José Mataix’Universidad de GranadaGranadaSpain
| | - Celia Rodríguez‐Pérez
- Departamento de Nutrición y Bromatología, Facultad de FarmaciaUniversidad de GranadaGranadaSpain
- Instituto de Nutrición y Tecnología de los Alimentos ‘José Mataix’Universidad de GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
| | - Angela Caunii
- “Victor Babes” University of Medicine and PharmacyTimisoaraRomania
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from TimisoaraTimisoaraRomania
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UKUniversity of GreenwichKentUK
| | | |
Collapse
|
7
|
Guo H, Wang Z, Yin K, Ma R, Zhang Y, Yin F, Li H, Yin D. Sciellin promotes the development and progression of thyroid cancer through the JAK2/STAT3 signaling pathway. Mol Carcinog 2024; 63:701-713. [PMID: 38411346 DOI: 10.1002/mc.23682] [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: 08/21/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 02/28/2024]
Abstract
Thyroid cancer (TC) is one of the most common endocrine tumors worldwide. Sciellin (SCEL) is involved in various disease processes, including burn wound healing and neutrophil extracellular traps (NETs); it is highly expressed in TC. However, its biological impact on TC and related mechanisms remain unclear. This study aimed to investigate the effect of SCEL on the function of human TC cell lines B-CPAP and OCUT-2C (cancer cell lines with BRAF V600E mutations). Analyses of data sets and clinical samples revealed enhanced expression of SCEL in TC than in adjacent normal tissue. SCEL knockout suppresses proliferation and cell cycle progression in TC cells, and these results were reversed by the upregulated SCEL expression in TC. SCEL knockout inhibited tumor development in xenograft mouse models. Western blot (WB) demonstrated that the expression of p-JAK2 and p-STAT3 was reduced in SCEL-knockdown TC. These results suggest that SCEL plays a key role in TC progression through the JAK2-STAT3 pathway. Therefore, SCEL can be considered a potential diagnostic biomarker and therapeutic target for TC.
Collapse
Affiliation(s)
- Haohao Guo
- Department of Thyroid Surgery, Zhengzhou, Henan, China
- Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
- Key Medicine Laboratory of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
| | - Ziyang Wang
- Department of Thyroid Surgery, Zhengzhou, Henan, China
| | - Keyu Yin
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Runsheng Ma
- Department of Thyroid Surgery, Zhengzhou, Henan, China
| | - Yifei Zhang
- Department of Thyroid Surgery, Zhengzhou, Henan, China
- Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
- Key Medicine Laboratory of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
| | - Fanxiang Yin
- Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hongqiang Li
- Department of Thyroid Surgery, Zhengzhou, Henan, China
| | - Detao Yin
- Department of Thyroid Surgery, Zhengzhou, Henan, China
- Engineering Research Center of Multidisciplinary Diagnosis and Treatment of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
- Key Medicine Laboratory of Thyroid Cancer of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Teng Y, Li Y, Wang L, Wang B, Su S, Chen J, Lu Z, Zhu H, Zhao M. Effectiveness and pharmacological mechanisms of Chinese herbal medicine for coronary heart disease complicated with heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117605. [PMID: 38128892 DOI: 10.1016/j.jep.2023.117605] [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: 02/15/2023] [Revised: 11/30/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine (CHM) is widely used for treating coronary heart disease complicated with heart failure (CHD-HF). However, the exact mechanisms involved are still not fully understood. AIM OF THE STUDY To assess the clinical effectiveness and potential pharmacological mechanisms of CHM for treating CHD-HF. METHODS Eight databases were retrieved for Randomized Controlled Trials of CHM for CHD-HF published from their inception to March 2023. Quality assessment of include studies was performed by the Cochrane risk-of-bias. Meta-analysis was used to assess the effectiveness of CHM for CHD-HF, and then core drugs and active ingredients were selected by data mining and network pharmacology. Finally, cluster and enrichment analysis were adopted to explore the potential targets and signaling pathways. RESULTS A total of 52 studies enrolling 5216 patients were included. Meta-analysis revealed that CHM treatment groups significantly improved left ventricular ejection fraction (LVEF), 6-min walk test (6-MWT), left ventricular end-diastolic dimension (LVEDD) and left ventricular end systolic diameter (LVESD) than control groups: [LVEF: SMD = 0.7, 95%CI (0.54, 0.87), p < 0.00001, I2 = 80%; 6-MWT: SMD = 0.72, 95%CI (0.58, 0.86), p < 0.0001, I2 = 67%; LVEDD: SMD = -0.79, 95%CI (-0.89, -0.69), p < 0.0001, I2 = 49%; LVESD: SMD = -0.6 (-0.74, -0.46), p < 0.0001, I2 = 0%]. The results of various biological information analysis showed the internal relationship between prescriptions, core drugs, active ingredients and therapeutic targets. Twelve core herbs with the most commonly use and high correlation were selected from 110 CHMs of 52 prescriptions for CHD-HF treatment, and further 65 effective components were screened out according to the most strength value, which were divided into 12 compounds such as terpenoids, flavonoids, steroids and alkaloids and etc. At the same time, 67 therapeutic targets of active ingredients in CHD-HF were filtrated. On these bases, cluster and enrichment analysis of the components and targets were used to explore relevant pharmacological mechanisms, mainly including anti-myocardial cell damage, anti-inflammation, anti-apoptosis, anti-fibrosis, regulation of oxidative stress, anticoagulation and angiogenesis, and improvement of glucose and fatty acid metabolism. CONCLUSION CHM are effective in treating CHD-HF compared with conventional treatment. Some of the included studies have high risks in the implementation of blinding, so more high-quality studies are needed. The active ingredients of CHM could protect the myocardium and improve pathological environment of CHD-HF in various ways. And CHM has the advantage of multi-component and multi-target treatment for complex diseases.
Collapse
Affiliation(s)
- Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Baofu Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jiaxin Chen
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Haiyan Zhu
- Department of Cardiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute of Cardiovascular Diseases, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
9
|
Khan F, Pandey P, Verma M, Upadhyay TK. Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies. Biomolecules 2024; 14:200. [PMID: 38397437 PMCID: PMC10886526 DOI: 10.3390/biom14020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer has become one of the most multifaceted and widespread illnesses affecting human health, causing substantial mortality at an alarming rate. After cardiovascular problems, the condition has a high occurrence rate and ranks second in terms of mortality. The development of new drugs has been facilitated by increased research and a deeper understanding of the mechanisms behind the emergence and advancement of the disease. Numerous preclinical and clinical studies have repeatedly demonstrated the protective effects of natural terpenoids against a range of malignancies. Numerous potential bioactive terpenoids have been investigated in natural sources for their chemopreventive and chemoprotective properties. In practically all body cells, the signaling molecule referred to as signal transducer and activator of transcription 3 (STAT3) is widely expressed. Numerous studies have demonstrated that STAT3 regulates its downstream target genes, including Bcl-2, Bcl-xL, cyclin D1, c-Myc, and survivin, to promote the growth of cells, differentiation, cell cycle progression, angiogenesis, and immune suppression in addition to chemotherapy resistance. Researchers viewed STAT3 as a primary target for cancer therapy because of its crucial involvement in cancer formation. This therapy primarily focuses on directly and indirectly preventing the expression of STAT3 in tumor cells. By explicitly targeting STAT3 in both in vitro and in vivo settings, it has been possible to explain the protective effect of terpenoids against malignant cells. In this study, we provide a complete overview of STAT3 signal transduction processes, the involvement of STAT3 in carcinogenesis, and mechanisms related to STAT3 persistent activation. The article also thoroughly summarizes the inhibition of STAT3 signaling by certain terpenoid phytochemicals, which have demonstrated strong efficacy in several preclinical cancer models.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India;
| | - Pratibha Pandey
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
- Department of Chemistry, University Institute of Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara 391760, India;
| |
Collapse
|
10
|
Hu X, Huang S, Ye S, Jiang J. The Natural Product Oridonin as an Anticancer Agent: Current Achievements and Problems. Curr Pharm Biotechnol 2024; 25:655-664. [PMID: 37605407 DOI: 10.2174/1389201024666230821110116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 08/23/2023]
Abstract
Oridonin, an active diterpenoid isolated from traditional Chinese herbal medicine, has received a rising attention for its remarkable roles in cancer therapy. In recent years, increasing evidences have revealed that oridonin inhibits the occurrence and development of tumor cells through multiple mechanisms, including induction of apoptosis and autophagy, cell cycle arrest, and inhibition of angiogenesis as well as migration and invasion. In addition, several molecular signal targets have been identified, including ROS, EGFR, NF-κB, PI3K/Akt, and MAPK. In this paper, we review considerable knowledge about the molecular mechanisms and signal targets of oridonin, which has been studied in recent years. It is expected that oridonin may be developed as a novel anti-tumor herbal medicine in human cancer treatment.
Collapse
Affiliation(s)
- Xiangyan Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| | - Sisi Huang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Pharmacy School of Fudan University, Shanghai, 200032, P.R. China
| | - Shiying Ye
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| | - Jinhuan Jiang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, P.R. China
| |
Collapse
|
11
|
Li GQ, Gao SX, Wang FH, Kang L, Tang ZY, Ma XD. Anticancer mechanisms on pyroptosis induced by Oridonin: New potential targeted therapeutic strategies. Biomed Pharmacother 2023; 165:115019. [PMID: 37329709 DOI: 10.1016/j.biopha.2023.115019] [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: 05/11/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023] Open
Abstract
Pyroptosis is a type of inflammatory cell death that is triggered by the formation of pores on the cell membrane by gasdermin (GSDM) family proteins. This process activates inflammasomes and leads to the maturation and release of proinflammatory cytokines such as interleukin-1β (IL-1β) and interleukin-18 (IL-18). Pyroptosis, a form of programmed cell death, has been found to be associated with various biomolecules such as caspases, granzymes, non-coding RNA (lncRNA), reactive oxygen species (ROS), and NOD-like receptor protein 3 (NLRP3). These biomolecules have been shown to play a dual role in cancer by affecting cell proliferation, metastasis, and the tumor microenvironment (TME), resulting in both tumor promotion and anti-tumor effects. Recent studies have found that Oridonin (Ori) has anti-tumor effects by regulating pyroptosis through various pathways. Ori can inhibit pyroptosis by inhibiting caspase-1, which is responsible for activating pyroptosis of the canonical pathway. Additionally, Ori can inhibit pyroptosis by inhibiting NLRP3, which is responsible for activating pyroptosis of the noncanonical pathway. Interestingly, Ori can also activate pyroptosis by activating caspase-3 and caspase-8, which are responsible for activating pyroptosis of the emerging pathway; Ori has been found to be effective in inhibiting pyroptosis by blocking the action of perforin, which is responsible for facilitating the entry of granzyme into cells and activating pyroptosis. Additionally, Ori plays a crucial role in regulating pyroptosis by promoting the accumulation of ROS while inhibiting the ncRNA and NLRP3 pathways. It is worth noting that all of these pathways ultimately regulate pyroptosis by influencing the cleavage of GSDM, which is a key factor in the process. These studies concludes that Ori has extensive anti-cancer effects that are related to its potential regulatory function on pyroptosis. The paper summarizes several potential ways in which Ori participates in the regulation of pyroptosis, providing a reference for further study on the relationship between Ori, pyroptosis, and cancer.
Collapse
Affiliation(s)
- Guo Qiang Li
- Pharmacy school, Dalian Medical University, Dalian 116044, Liaoning, PR China
| | - Shi Xiang Gao
- Pharmacy school, Dalian Medical University, Dalian 116044, Liaoning, PR China
| | - Fu Han Wang
- Pharmacy school, Dalian Medical University, Dalian 116044, Liaoning, PR China
| | - Le Kang
- Department of Cardiac Surgery, Zhongshan Hospital, Affiliated Fudan University, Shang Hai 200030, PR China.
| | - Ze Yao Tang
- Pharmacy school, Dalian Medical University, Dalian 116044, Liaoning, PR China.
| | - Xiao Dong Ma
- Pharmacy school, Dalian Medical University, Dalian 116044, Liaoning, PR China.
| |
Collapse
|
12
|
Wu J, Nie Q, Li G, Zhu K. Identifying driver pathways based on a parameter-free model and a partheno-genetic algorithm. BMC Bioinformatics 2023; 24:211. [PMID: 37221474 DOI: 10.1186/s12859-023-05319-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/04/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Tremendous amounts of omics data accumulated have made it possible to identify cancer driver pathways through computational methods, which is believed to be able to offer critical information in such downstream research as ascertaining cancer pathogenesis, developing anti-cancer drugs, and so on. It is a challenging problem to identify cancer driver pathways by integrating multiple omics data. RESULTS In this study, a parameter-free identification model SMCMN, incorporating both pathway features and gene associations in Protein-Protein Interaction (PPI) network, is proposed. A novel measurement of mutual exclusivity is devised to exclude some gene sets with "inclusion" relationship. By introducing gene clustering based operators, a partheno-genetic algorithm CPGA is put forward for solving the SMCMN model. Experiments were implemented on three real cancer datasets to compare the identification performance of models and methods. The comparisons of models demonstrate that the SMCMN model does eliminate the "inclusion" relationship, and produces gene sets with better enrichment performance compared with the classical model MWSM in most cases. CONCLUSIONS The gene sets recognized by the proposed CPGA-SMCMN method possess more genes engaging in known cancer related pathways, as well as stronger connectivity in PPI network. All of which have been demonstrated through extensive contrast experiments among the CPGA-SMCMN method and six state-of-the-art ones.
Collapse
Affiliation(s)
- Jingli Wu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, China.
- Guangxi Key Lab of Multi-source Information Mining and Security, Guangxi Normal University, Guilin, China.
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, China.
| | - Qinghua Nie
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, China
- Guangxi Key Lab of Multi-source Information Mining and Security, Guangxi Normal University, Guilin, China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, China
| | - Gaoshi Li
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin, China
- Guangxi Key Lab of Multi-source Information Mining and Security, Guangxi Normal University, Guilin, China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, China
| | - Kai Zhu
- Guangxi Key Lab of Multi-source Information Mining and Security, Guangxi Normal University, Guilin, China
- College of Computer Science and Engineering, Guangxi Normal University, Guilin, China
| |
Collapse
|
13
|
Unraveling the function of epithelial-mesenchymal transition (EMT) in colorectal cancer: Metastasis, therapy response, and revisiting molecular pathways. Biomed Pharmacother 2023; 160:114395. [PMID: 36804124 DOI: 10.1016/j.biopha.2023.114395] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/03/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Colorectal cancer (CRC) is a dangerous form of cancer that affects the gastrointestinal tract. It is a major global health concern, and the aggressive behavior of tumor cells makes it difficult to treat, leading to poor survival rates for patients. One major challenge in treating CRC is the metastasis, or spread, of the cancer, which is a major cause of death. In order to improve the prognosis for patients with CRC, it is necessary to focus on ways to inhibit the cancer's ability to invade and spread. Epithelial-mesenchymal transition (EMT) is a process that is linked to the spread of cancer cells, also known as metastasis. The process transforms epithelial cells into mesenchymal ones, increasing their mobility and ability to invade other tissues. This has been shown to be a key mechanism in the progression of colorectal cancer (CRC), a particularly aggressive form of gastrointestinal cancer. The activation of EMT leads to increases in the spread of CRC cells, and during this process, levels of the protein E-cadherin decrease while levels of N-cadherin and vimentin increase. EMT also contributes to the development of resistance to chemotherapy and radiation therapy in CRC. Non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), play a role in regulating EMT in CRC, often through their ability to "sponge" microRNAs. Anti-cancer agents have been shown to suppress EMT and reduce the progression and spread of CRC cells. These findings suggest that targeting EMT or related mechanisms may be a promising approach for treating CRC patients in the clinic.
Collapse
|