1
|
Khatua S, Nandi S, Nag A, Sen S, Chakraborty N, Naskar A, Gürer ES, Calina D, Acharya K, Sharifi-Rad J. Homoharringtonine: updated insights into its efficacy in hematological malignancies, diverse cancers and other biomedical applications. Eur J Med Res 2024; 29:269. [PMID: 38704602 PMCID: PMC11069164 DOI: 10.1186/s40001-024-01856-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/13/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
HHT has emerged as a notable compound in the realm of cancer treatment, particularly for hematological malignancies. Its multifaceted pharmacological properties extend beyond traditional applications, warranting an extensive review of its mechanisms and efficacy. This review aims to synthesize comprehensive insights into the efficacy of HHT in treating hematological malignancies, diverse cancers, and other biomedical applications. It focuses on elucidating the molecular mechanisms, therapeutic potential, and broader applications of HHT. A comprehensive search for peer-reviewed papers was conducted across various academic databases, including ScienceDirect, Web of Science, Scopus, American Chemical Society, Google Scholar, PubMed/MedLine, and Wiley. The review highlights HHT's diverse mechanisms of action, ranging from its role in leukemia treatment to its emerging applications in managing other cancers and various biomedical conditions. It underscores HHT's influence on cellular processes, its efficacy in clinical settings, and its potential to alter pathological pathways. HHT demonstrates significant promise in treating various hematological malignancies and cancers, offering a multifaceted approach to disease management. Its ability to impact various physiological pathways opens new avenues for therapeutic applications. This review provides a consolidated foundation for future research and clinical applications of HHT in diverse medical fields.
Collapse
Affiliation(s)
- Somanjana Khatua
- Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, Uttar Pradesh, 211002, India
| | - Sudeshna Nandi
- Department of Botany, Molecular and Applied Mycology and Plant Pathology Laboratory, University of Calcutta, 35, Ballygung Circular Road, Kolkata, India
| | - Anish Nag
- Department of Life Sciences, CHRIST (Deemed to Be University), Bangalore Central Campus, Bangalore, Karnataka, India
| | - Surjit Sen
- Department of Botany, Fakir Chand College, Diamond Harbour, South 24-Parganas, Kolkata, India
| | | | - Arghya Naskar
- Department of Botany, Molecular and Applied Mycology and Plant Pathology Laboratory, University of Calcutta, 35, Ballygung Circular Road, Kolkata, India
| | - Eda Sönmez Gürer
- Department of Pharmacognosy, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Krishnendu Acharya
- Department of Botany, Molecular and Applied Mycology and Plant Pathology Laboratory, University of Calcutta, 35, Ballygung Circular Road, Kolkata, India.
| | | |
Collapse
|
2
|
Entezari M, Tayari A, Paskeh MDA, Kheirabad SK, Naeemi S, Taheriazam A, Dehghani H, Salimimoghadam S, Hashemi M, Mirzaei S, Samarghandian S. Curcumin in treatment of hematological cancers: Promises and challenges. J Tradit Complement Med 2024; 14:121-134. [PMID: 38481552 PMCID: PMC10927384 DOI: 10.1016/j.jtcme.2023.10.004] [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] [Academic Contribution Register] [Received: 03/31/2023] [Revised: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2024] Open
Abstract
Hematological cancers include leukemia, myeloma and lymphoma and up to 178.000 new cases are diagnosed with these tumors each year. Different kinds of treatment including radiotherapy, chemotherapy, immunotherapy and stem cell transplantation have been employed in the therapy of hematological cancers. However, they are still causing death among patients. On the other hand, curcumin as an anti-cancer agent for the suppression of human cancers has been introduced. The treatment of hematological cancers using curcumin has been followed. Curcumin diminishes viability and survival rate of leukemia, myeloma and lymphoma cells. Curcumin stimulates apoptosis and G2/M arrest to impair progression of tumor. Curcumin decreases levels of matrix metalloproteinases in suppressing cancer metastasis. A number of downstream targets including VEGF, Akt and STAT3 undergo suppression by curcumin in suppressing progression of hematological cancers. Curcumin stimulates DNA damage and reduces resistance of cancer cells to irradiation. Furthermore, curcumin causes drug sensitivity of hematological tumors, especially myeloma. For targeted delivery of curcumin and improving its pharmacokinetic and anti-cancer features, nanostructures containing curcumin and other anti-cancer agents have been developed.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Armita Tayari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Simin Khorsand Kheirabad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sahar Naeemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Dehghani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Medical Laboratory Sciences, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
3
|
Kozalak G, Koşar A. Autophagy-related mechanisms for treatment of multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:838-857. [PMID: 38239705 PMCID: PMC10792488 DOI: 10.20517/cdr.2023.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 09/15/2023] [Revised: 12/12/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Multiple myeloma (MM) is a type of hematological cancer that occurs when B cells become malignant. Various drugs such as proteasome inhibitors, immunomodulators, and compounds that cause DNA damage can be used in the treatment of MM. Autophagy, a type 2 cell death mechanism, plays a crucial role in determining the fate of B cells, either promoting their survival or inducing cell death. Therefore, autophagy can either facilitate the progression or hinder the treatment of MM disease. In this review, autophagy mechanisms that may be effective in MM cells were covered and evaluated within the contexts of unfolded protein response (UPR), bone marrow microenvironment (BMME), drug resistance, hypoxia, DNA repair and transcriptional regulation, and apoptosis. The genes that are effective in each mechanism and research efforts on this subject were discussed in detail. Signaling pathways targeted by new drugs to benefit from autophagy in MM disease were covered. The efficacy of drugs that regulate autophagy in MM was examined, and clinical trials on this subject were included. Consequently, among the autophagy mechanisms that are effective in MM, the most suitable ones to be used in the treatment were expressed. The importance of 3D models and microfluidic systems for the discovery of new drugs for autophagy and personalized treatment was emphasized. Ultimately, this review aims to provide a comprehensive overview of MM disease, encompassing autophagy mechanisms, drugs, clinical studies, and further studies.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabancı University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
4
|
Liao X, Cai D, Liu J, Hu H, You R, Pan Z, Chen S, Xu K, Dai W, Zhang S, Lin X, Huang H. Deletion of Mettl3 in mesenchymal stem cells promotes acute myeloid leukemia resistance to chemotherapy. Cell Death Dis 2023; 14:796. [PMID: 38052820 PMCID: PMC10698052 DOI: 10.1038/s41419-023-06325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2023] [Revised: 11/10/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Acute myeloid leukemia (AML) cell survival and chemoresistance are influenced by the existence of bone marrow mesenchymal stem cells (BMMSCs); however, the pathways by which BMMSCs contribute to these processes remain unclear. We earlier revealed that methyltransferase-like 3 (METTL3) expression is significantly reduced in AML BMMSCs and that METTL3 mediates BMMSC adipogenesis to promote chemoresistance in human AML cell lines in vitro. In this investigation, we evaluated the METTL3 function in vivo. Mice exhibiting a conditional removal of Mettl3 in BMMSCs were developed by mating Prrx1-CreERT2;Mettl3fl/+ mice with Mettl3fl/fl mice using the CRISPR-Cas9 system. The Mettl3 deletion increased bone marrow adiposity, enhanced disease progression in the transplantation-induced MLL-AF9 AML mouse model, and chemoresistance to cytarabine. The removal of Mettl3 in BMMSCs resulted in a significant increase in BMMSC adipogenesis. This effect was attributed to the downregulation of AKT1 expression, an AKT serine/threonine kinase 1, in an m6A-dependent manner. The development of chemoresistance in AML is linked to the promoted adipogenesis of BMMSCs. We conclude that METTL3 expression in BMMSCs has a critical function in limiting AML progression and chemoresistance, providing a basis for the progression of therapeutic approaches for AML.
Collapse
Affiliation(s)
- Xinai Liao
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Danni Cai
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Haoran Hu
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Ruolan You
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Zhipeng Pan
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Shucheng Chen
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Kaiming Xu
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Wei Dai
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Shuxia Zhang
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, 350122, Fuzhou, Fujian, China.
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, 350001, Fuzhou, Fujian, China.
| |
Collapse
|
5
|
Wang L, Yu T, Dong F, Xu J, Fu J, Sun H. Tongqiao Mingmu formula alleviates retinal ganglion cell autophagy through PI3K/AKT/mTOR pathway. Anat Rec (Hoboken) 2023; 306:3120-3130. [PMID: 36098527 DOI: 10.1002/ar.25060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/19/2022] [Revised: 06/20/2022] [Accepted: 07/12/2022] [Indexed: 11/06/2022]
Abstract
Glaucoma is a severe blindness-causing optic nerve disease characterized by a loss of retinal ganglion cells (RGCs). Previous studies have shown that the Tongqiao Mingmu (TQMM) formula can reduce retinal and optic nerve damage, but its mechanism of action requires further elucidation. In this study, an RGC injury model was prepared using glutamate and then treated with serum-containing drug from the TQMM formula (hereafter called "TQMM formula serum"). In the glutamate-induced RGC injury model, cell viability decreased with an increase in glutamate concentration, whereas the expression of autophagy-related biomarkers LC3 and Belicin-1 increased. An adenovirus transfection experiment revealed that glutamate markedly promoted autophagic flux in RGCs. Notably, TQMM formula serum inhibited the expression of autophagy-related biomarkers, reduced autophagy flux, and reversed the damage caused by glutamate to RGCs. Furthermore, the PI3K inhibitor LY294002 was used to intervene in the RGC autophagy model and was found to suppress the PI3K/AKT/mTOR pathway and enhance RGC autophagy. However, TQMM formula serum could generate an opposite effect and upregulate the expressions of the PI3K/AKT/mTOR pathway genes and proteins. In conclusion, the TQMM formula can prevent glutamate-induced autophagy in RGCs, possibly by activating the PI3K/AKT/mTOR pathway and reducing the expression of autophagy-related biomarkers LC3 and Belicin-1 to attenuate autophagy and maintain RGC viability.
Collapse
Affiliation(s)
- Liyuan Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Ophthalmology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- Heilongjiang Academy of Sciences of Traditional Chinese Medicine, Harbin, China
| | - Tianyang Yu
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Acupuncture, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Feixue Dong
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Ophthalmology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiayu Xu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jin Fu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - He Sun
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Ophthalmology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Islam R, Yan MP, Yen KP, Rasol NE, Meng CK, Wai LK. Synthesis and biological evaluation of chromone derivatives against triple-negative breast cancer cells. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03048-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/19/2023]
|
7
|
Qu M, Li J, Yuan L. Uncovering the action mechanism of homoharringtonine against colorectal cancer by using network pharmacology and experimental evaluation. Bioengineered 2021; 12:12940-12953. [PMID: 34847838 PMCID: PMC8810123 DOI: 10.1080/21655979.2021.2012626] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/06/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 02/07/2023] Open
Abstract
Homoharringtonine (HHT), an Food and Drug Administration (FDA)-approved anti-leukemia drug, exerts anti-tumor activity in several solid tumors, including colorectal cancer (CRC). However, its mechanism of action in CRC progression has not been comprehensively elucidated. The drug-disease targets were obtained using publicly available databases. Protein-protein interaction (PPI) network, Gene ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were performed to reveal the core targets, biological processes and signaling pathways of HHT against CRC. Cell and animal experiments were performed to validate the inhibitory effects of HHT on CRC. A total of 98 overlapping target genes of HHT and CRC were predicted. Through PPI network and topology analysis, we screened out 23 hub genes. Enrichment assays showed 163 biological processes (BP), 18 cell components (CC), 35 molecular functions (MF), and 85 related pathways. Functionally, HHT inhibited CRC cell proliferation, cell cycle progression, colony formation, migration and invasion, and promoted apoptosis. HHT treatment resulted in the inactivation of PI3K/AKT/mTOR signaling in CRC cells. Moreover, activation of PI3K/AKT/mTOR signaling by 740Y-P abated the suppressive effects of HHT on cell malignant phenotypes. Furthermore, HHT repressed CRC tumor growth in nude mice. Our current study demonstrated that HHT repressed CRC progression at least partly by inactivating PI3K/AKT/mTOR signaling pathways, highlighting HHT as a potential therapeutic agent for CRC patients.
Collapse
Affiliation(s)
- Muwen Qu
- Department of Anorectal Branch, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junyi Li
- Department of Surgery, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lingling Yuan
- Department of Dermatology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Pan ZP, Wang B, Hou DY, You RL, Wang XT, Xie WH, Huang HF. METTL3 mediates bone marrow mesenchymal stem cell adipogenesis to promote chemoresistance in acute myeloid leukaemia. FEBS Open Bio 2021; 11:1659-1672. [PMID: 33932138 PMCID: PMC8167861 DOI: 10.1002/2211-5463.13165] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/17/2021] [Revised: 03/13/2021] [Accepted: 04/12/2021] [Indexed: 02/02/2023] Open
Abstract
Adipogenesis of bone marrow mesenchymal stem cells (MSCs) promotes chemoresistance of acute myeloid leukaemia (AML) cells. MSCs from AML patients (AML‐MSCs) display enhanced adipogenesis compared with bone marrow MSCs from healthy donors. However, the precise molecular mechanism by which adipogenesis of MSCs from AML marrow differs from normal counterparts remains obscure. We found that METTL3 significantly inhibits MSC adipogenesis. Here, we aimed to identify the molecular mechanism linking METTL3 and MSC adipogenesis. Analysis of m6A epigenetic changes in MSCs determined via RIP‐qPCR and MeRIP‐qPCR indicated that METTL3 affects AKT protein expression in MSCs by mediating m6A modification of AKT1‐mRNA. Downregulated METTL3 expression in AML‐MSCs induced an increase in AKT protein, resulting in enhanced MSC adipogenesis, thereby contributing to chemoresistance in AML cells. Therefore, targeting AKT regulation by mRNA modification in MSC adipogenesis might provide a novel therapeutic strategy to overcome AML chemoresistance.
Collapse
Affiliation(s)
- Zhi-Peng Pan
- Central Laboratory, Fujian Medical University Union Hospital, China
| | - Bin Wang
- Central Laboratory, Fujian Medical University Union Hospital, China.,Clinical Laboratory, Fujian Maternal and Child Health Hospital, Fujian Children's Hospital, China
| | - Di-Yu Hou
- Central Laboratory, Fujian Medical University Union Hospital, China
| | - Ruo-Lan You
- Central Laboratory, Fujian Medical University Union Hospital, China
| | - Xiao-Ting Wang
- Central Laboratory, Fujian Medical University Union Hospital, China
| | - Wen-Hui Xie
- Graduate School, Fujian Medical University, Fujian Medical University Union Hospital, China
| | - Hui-Fang Huang
- Central Laboratory, Fujian Medical University Union Hospital, China
| |
Collapse
|
9
|
Abstract
Cephalotaxus is the only genus of Cephalotaxaceae family, and its natural resources are declining due to habitat fragmentation, excessive exploitation and destruction. In many areas of China, folk herbal doctors traditionally use Cephalotaxus plants to treat innominate swollen poison, many of which are cancer. Not only among Han people, but also among minority ethnic groups, Cephalotaxus is used to treat various diseases, e.g., cough, internal bleeding and cancer in Miao medicine, bruises, rheumatism and pain in Yao medicine, and ascariasis, hookworm disease, scrofula in She medicine, etc. Medicinal values of some Cephalotaxus species and compounds are acknowledged officially. However, there is a lack of comprehensive review summarizing the ethnomedicinal knowledge of Cephalotaxus, relevant medicinal phytometabolites and their bioactivities. The research progresses in ethnopharmacology, chemodiversity, and bioactivities of Cephalotaxus medicinal plants are reviewed and commented here. Knowledge gaps are pinpointed and future research directions are suggested. Classic medicinal books, folk medicine books, herbal manuals and ethnomedicinal publications were reviewed for the genus Cephalotaxus (Sanjianshan in Chinese). The relevant data about ethnobotany, phytochemistry, and pharmacology were collected as comprehensively as possible from online databases including Scopus, NCBI PubMed, Bing Scholar, and China National Knowledge Infrastructure (CNKI). "Cephalotaxus", and the respective species name were used as keywords in database search. The obtained articles of the past six decades were collated and analyzed. Four Cephalotaxus species are listed in the official medicinal book in China. They are used as ethnomedicines by many ethnic groups such as Miao, Yao, Dong, She and Han. Inspirations are obtained from traditional applications, and Cephalotaxus phytometabolites are developed into anticancer reagents. Cephalotaxine-type alkaloids, homoerythrina-type alkaloids and homoharringtonine (HHT) are abundant in Cephalotaxus, e.g., C. lanceolata, C. fortunei var. alpina, C. griffithii, and C. hainanensis, etc. New methods of alkaloid analysis and purification are continuously developed and applied. Diterpenoids, sesquiterpenoids, flavonoids, lignans, phenolics, and other components are also identified and isolated in various Cephalotaxus species. Alkaloids such as HHT, terpenoids and other compounds have anticancer activities against multiple types of human cancer. Cephalotaxus extracts and compounds showed anti-inflammatory and antioxidant activities, immunomodulatory activity, antimicrobial activity and nematotoxicity, antihyperglycemic effect, and bone effect, etc. Drug metabolism and pharmacokinetic studies of Cephalotaxus are increasing. We should continue to collect and sort out folk medicinal knowledge of Cephalotaxus and associated organisms, so as to obtain new enlightenment to translate traditional tips into great therapeutic drugs. Transcriptomics, genomics, metabolomics and proteomics studies can contribute massive information for bioactivity and phytochemistry of Cephalotaxus medicinal plants. We should continue to strengthen the application of state-of-the-art technologies in more Cephalotaxus species and for more useful compounds and pharmacological activities.
Collapse
|
10
|
Wang LB, Wang DN, Wu LG, Cao J, Tian JH, Liu R, Ma R, Yu JJ, Wang J, Huang Q, Xiong WY, Zhang X. Homoharringtonine inhibited breast cancer cells growth via miR-18a-3p/AKT/mTOR signaling pathway. Int J Biol Sci 2021; 17:995-1009. [PMID: 33867824 PMCID: PMC8040299 DOI: 10.7150/ijbs.44907] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/13/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Homoharringtonine (HHT), a natural alkaloid derived from the cephalotaxus, exhibited its anti-cancer effects in hematological malignancies clinically. However, its pesticide effects and mechanisms in treating solid tumors remain unclear. In this study, we found that HHT was capable of inhibiting tumor growth after 5-days treatment of breast cancer cells, MCF-7, in vivo. Furthemore, HHT also significantly inhibited the cancer cell growth and induced cell apoptosis in vitro. miRNA sequencing proved miR-18a-3p was noticeably downregulated in the cells after HHT treatment. Moreover, downregulating miR-18a-3p increased HHT-induced cell apoptosis; our data supported that HHT suppressed miR-18a-3p expression and inhibited tumorigenesis might via AKT-mTOR signaling pathway. In conclusion: our study proved that HHT suppressed breast cancer cell growth and promoted apoptosis mediated by regulating of the miR-18a-3p-AKT-mTOR signaling pathway, HHT may be a promising antitumor agent in breast cancer treatment.
Collapse
Affiliation(s)
- Li-Bin Wang
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Dan-Ni Wang
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China.,Gansu Provincial Hospital, Clinical Laboratory Center, Lanzhou, 730000, China
| | - Li-Gang Wu
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Jia Cao
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Jin-Hai Tian
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Rong Liu
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Rong Ma
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Jing-Jing Yu
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Jia Wang
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Qi Huang
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| | - Wen-Yong Xiong
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China.,Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan University, Kunming, 650091, China
| | - Xu Zhang
- The General Hospital of Ningxia Medical University, Biochip Research Center, Yinchuan, 750001, China
| |
Collapse
|
11
|
Zhou X, Wang X, Zhu H, Gu G, Zhan Y, Liu C, Sun G. PI3K inhibition sensitizes EGFR wild-type NSCLC cell lines to erlotinib chemotherapy. Exp Ther Med 2020; 21:9. [PMID: 33235618 PMCID: PMC7678642 DOI: 10.3892/etm.2020.9441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/23/2019] [Accepted: 01/14/2020] [Indexed: 12/30/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) bring significant benefits for patients with cancers harboring epidermal growth factor receptor (EGFR) mutations. However, after treatment for a certain period, most patients ultimately acquire resistance. Numerous studies indicated that PI3K has an important role in tumor cell growth and drug sensitivity. Furthermore, inhibition of PI3K may lead to sensitization of non-small cell lung cancer (NSCLC) cells to EGFR-TKIs. The aim of the present study was to explore whether LY294002, an inhibitor of PI3K, is able to improve the sensitivity of NSCLC cell lines with wild-type EGFR to the EGFR-TKI erlotinib. An MTT assay was used to examine the effect of combined treatment with LY294002 and erlotinib on cell survival of two EGFR wild-type NSCLC cell lines, NCI-H661 and NCI-H460. Furthermore, flow cytometry was used to assess apoptosis in NCI-H661 and NCI-H460 cells after treatment with erlotinib and LY294002. In addition, the expression of downstream proteins was detected by western blot analysis. The results indicated that the number of viable NCI-H661 and NCI-H460 cells was dose-dependently reduced by erlotinib or LY294002. Compared to treatment with erlotinib alone, the cell apoptosis was enhanced if combined treatment of erlotinib and LY294002 was performed in NCI-H661 cells. Furthermore, combination treatment of erlotinib and LY294002 resulted in a significant reduction of phosphorylated p70S6K levels in NCI-H661 [PI3K catalytic subunit alpha (PI3KCA) wild-type] cells. However, this phenomenon was not observed in the NCI-H460 cell line (PIK3CA mutant-type). In conclusion, the present study indicated that inhibition of PI3K may have the potential to improve the sensitivity of NSCLC cells to an EGFR-TKI. However, the therapeutic effect may depend on the mutation status of PIK3CA.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Second Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Xiaowen Wang
- Department of Second Breast Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Hongge Zhu
- Department of Second Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Guomin Gu
- Department of Second Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Yiyi Zhan
- Department of Second Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Chunling Liu
- Department of Second Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Gang Sun
- Department of Breast, Head and Neck Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
12
|
Hou D, Wang B, You R, Wang X, Liu J, Zhan W, Chen P, Qin T, Zhang X, Huang H. Stromal cells promote chemoresistance of acute myeloid leukemia cells via activation of the IL-6/STAT3/OXPHOS axis. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1346. [PMID: 33313091 PMCID: PMC7723653 DOI: 10.21037/atm-20-3191] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 01/08/2023]
Abstract
Background Bone marrow stromal cells (BMSCs) are known to promote chemoresistance in acute myeloid leukemia (AML) cells. However, the molecular basis for BMSC-associated AML chemoresistance remains largely unexplored. Methods The mitochondrial oxidative phosphorylation (OXPHOS) levels of AML cells were measured by a Seahorse XFe24 cell metabolic analyzer. The activity of total or mitochondrial signal transducer and transcription activator 3 (STAT3) in AML cells was explored by flow cytometry and Western blotting. Real-time quantitative PCR, Western blotting and enzyme-linked immunosorbent assay (ELISA) were used to analyze expression of interleukin 6 (IL-6) in the human BMSC line HS-5, and IL-6 was knocked out in HS-5 cells by CRISPR/Cas9 system. Results In this study, we observed that co-culturing with BMSCs heightened OXPHOS levels in AML cells, thus promoting chemoresistance in these cells. HS-5 cell-induced upregulation of OXPHOS is dependent on the activation of STAT3, especially on that of mitochondrial serine phosphorylated STAT3 (pS-STAT3) in AML cells. The relationship among pS-STAT3, OXPHOS, and chemosensitivity of AML cells induced by BMSCs was demonstrated by the STAT3 activator and inhibitor, which upregulated and downregulated the levels of mitochondrial pS-STAT3 and OXPHOS, respectively. Intriguingly, AML cells remodeled HS-5 cells to secrete more IL-6, which augmented mitochondrial OXPHOS in AML cells and stimulated their chemoresistance. IL-6 knockout in HS-5 cells impaired the ability of these cells to activate STAT3, to increase OXPHOS, or to promote chemoresistance in AML cells. Conclusions BMSCs promoted chemoresistance in AML cells via the activation of the IL-6/STAT3/OXPHOS pathway. These findings exhibit a novel mechanism of chemoresistance in AML cells in the bone marrow microenvironment from a metabolic perspective.
Collapse
Affiliation(s)
- Diyu Hou
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bin Wang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China.,Clinical Laboratory, Fujian Children's Hospital, Fuzhou, China
| | - Ruolan You
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoting Wang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jingru Liu
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Weiwu Zhan
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Tiandi Qin
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xuehao Zhang
- School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
| | - Huifang Huang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
13
|
Walker ZJ, Idler BM, Davis LN, Stevens BM, VanWyngarden MJ, Ohlstrom D, Bearrows SC, Hammes A, Smith CA, Jordan CT, Mark TM, Forsberg PA, Sherbenou DW. Exploiting Protein Translation Dependence in Multiple Myeloma with Omacetaxine-Based Therapy. Clin Cancer Res 2020; 27:819-830. [PMID: 33109736 DOI: 10.1158/1078-0432.ccr-20-2246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2020] [Revised: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The prognosis of patients with multiple myeloma who are resistant to proteasome inhibitors, immunomodulatory drugs (IMiD), and daratumumab is extremely poor. Even B-cell maturation antigen-specific chimeric antigen receptor T-cell therapies provide only a temporary benefit before patients succumb to their disease. In this article, we interrogate the unique sensitivity of multiple myeloma cells to the alternative strategy of blocking protein translation with omacetaxine. EXPERIMENTAL DESIGN We determined protein translation levels (n = 17) and sensitivity to omacetaxine (n = 51) of primary multiple myeloma patient samples. Synergy was evaluated between omacetaxine and IMiDs in vitro, ex vivo, and in vivo. Underlying mechanism was investigated via proteomic analysis. RESULTS Almost universally, primary patient multiple myeloma cells exhibit >2.5-fold increased rates of protein translation compared with normal marrow cells. Ex vivo treatment with omacetaxine resulted in >50% reduction in viable multiple myeloma cells. In this cohort, high levels of translation serve as a biomarker for patient multiple myeloma cell sensitivity to omacetaxine. Unexpectedly, omacetaxine demonstrated synergy with IMiDs in multiple myeloma cell lines in vitro. In addition, in an IMiD-resistant relapsed patient sample, omacetaxine/IMiD combination treatment resensitized the multiple myeloma cells to the IMiD. Proteomic analysis found that the omacetaxine/IMiD combination treatment produced a double-hit on the IRF4/c-MYC pathway, which is critical to multiple myeloma survival. CONCLUSIONS Overall, protein translation inhibitors represent a potential new drug class for myeloma treatment and provide a rationale for conducting clinical trials with omacetaxine alone and in combination with IMiDs for patients with relapsed/refractory multiple myeloma.
Collapse
Affiliation(s)
- Zachary J Walker
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Beau M Idler
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lorraine N Davis
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael J VanWyngarden
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Denis Ohlstrom
- Biomedical Sciences and Biotechnology, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shelby C Bearrows
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Andrew Hammes
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Clayton A Smith
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tomer M Mark
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter A Forsberg
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado. .,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
14
|
Wang B, Wang X, Hou D, Huang Q, Zhan W, Chen C, Liu J, You R, Xie J, Chen P, Huang H. Exosomes derived from acute myeloid leukemia cells promote chemoresistance by enhancing glycolysis-mediated vascular remodeling. J Cell Physiol 2018; 234:10602-10614. [PMID: 30417360 DOI: 10.1002/jcp.27735] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2018] [Accepted: 10/18/2018] [Indexed: 12/20/2022]
Abstract
Acute myeloid leukemia (AML) is the most common type of leukemia in adults. AML cells secrete angiogenic factors to remodel vasculature and acquire chemoresistance; however, antiangiogenic drugs are often ineffective in AML treatment. Cancer cell-derived exosomes can induce angiogenesis, but their role in vascular remodeling during AML is unclear. Here, we found that exosomes secreted by AML cells promoted proliferation and migration and tube-forming activity of human umbilical vein endothelial cells (HUVECs), whereas HUVECs conferred chemoresistance to AML cells. AML cell-derived exosomes contained vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) messenger RNA and induced VEGFR expression in HUVECs. Furthermore, they enhanced glycolysis, which correlated with HUVEC proliferation, tube formation, and resistance to apoptosis. Thus, AML cells secrete VEGF/VEGFR-containing exosomes that induce glycolysis in HUVECs leading to vascular remodeling and acquisition of chemoresistance. These findings may contribute to the development of novel therapeutic strategies targeting exosomes in AML.
Collapse
Affiliation(s)
- Bin Wang
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoting Wang
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Diyu Hou
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qian Huang
- Department of Hepatobiliary Disease, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Weiwu Zhan
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Canwei Chen
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China.,Department of Clinical Laboratory, The Hospital of Nanan City, Nanan, China
| | - Jingru Liu
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ruolan You
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jieqiong Xie
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Chen
- Department of Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huifang Huang
- Department of Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
15
|
Zhang Q, Wang C, Han X, Yang G, Ge Z, Zhang G. Knockdown of ADAM17 inhibits cell proliferation and increases oxaliplatin sensitivity in HCT-8 colorectal cancer through EGFR-PI3K-AKT activation. Biochem Biophys Res Commun 2018; 503:2333-2339. [DOI: 10.1016/j.bbrc.2018.06.158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2018] [Accepted: 06/27/2018] [Indexed: 11/25/2022]
|