1
|
Panyajai P, Viriyaadhammaa N, Chiampanichayakul S, Sakamoto Y, Okonogi S, Moroishi T, Anuchapreeda S. Anticancer and cancer preventive activities of shogaol and curcumin from Zingiberaceae family plants in KG-1a leukemic stem cells. BMC Complement Med Ther 2025; 25:87. [PMID: 40022126 PMCID: PMC11869560 DOI: 10.1186/s12906-025-04829-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Leukemic stem cells (LSCs) present a significant challenge in the treatment of leukemia in patients because they exhibit a drug-resistant phenotype, making them difficult to eliminate. Searching for a new anticancer drug is crucial for improving leukemia treatment. Plants from the Zingiberaceae family are frequently used in traditional medicines due to their safety and accessibility. This study explores the anticancer activity, cancer preventive properties, and apoptosis inducing mechanisms of active compounds derived from these plants. METHODS Ten crude ethanolic extracts from each plant of the Zingiberaceae family were obtained using maceration techniques. The cytotoxicity of all extracts anticancer was assessed in comparison to anticancer drugs (cyclophosphamide, cytarabine, doxorubicin, and idarubicin) using MTT assay on cancer cell lines (KG-1a, K562, A549, MCF-7, and HeLa) and peripheral blood mononuclear cells (PBMCs). Cancer prevention properties of the effective extracts and their active compounds were evaluated by measuring the levels of tumor necrosis factor-alpha (TNF-α), interleukin-2 (IL-2), and nitric oxide (NO) using commercial kits. Cell cycle and cell death analyses were conducted using flow cytometry. Moreover, the effects of effective extracts and their active compounds on WT1 and CD34 expressions, as well as the apoptosis mechanism induced by the active compounds in KG-1a cells, were determined by Western blotting. RESULTS The cytotoxicity tests revealed that crude ethanolic extracts from Curcuma longa, C. zedoaria, and Zingiber officinale exhibited effective cytotoxicity against cancer cell lines while demonstrating lower impact on PBMCs. The active compounds of C. longa and C. zedoaria are curcuminoids, while those in Z. officinale are shogaol and gingerol. Notably, the IC20 values of curcuminoids and shogaol exhibited cancer prevention properties and reduced WT1 protein expression, thereby inhibiting cell proliferation. Furthermore, shogaol and curcumin demonstrated the ability to arrest the cell cycle at the G2/M phase and induce apoptosis through the Akt pathway. CONCLUSION These findings highlight shogaol and curcumin as promising compounds for leukemia treatment.
Collapse
Affiliation(s)
- Pawaret Panyajai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natsima Viriyaadhammaa
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Yasuhisa Sakamoto
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Siriporn Okonogi
- Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Toshiro Moroishi
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, Kumamoto, 860-8556, Japan.
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
2
|
Kong R, Shi J, Xie K, Wu H, Wang X, Zhang Y, Wang Y. A Study of JUN's Promoter Region and Its Regulators in Chickens. Genes (Basel) 2024; 15:1351. [PMID: 39457475 PMCID: PMC11508107 DOI: 10.3390/genes15101351] [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: 09/16/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Background: The Jun proto-oncogene (JUN), also referred to as C-JUN, is an integral component of the JNK signaling pathway, which is crucial for the formation and differentiation of spermatogonial stem cells (SSCs). Investigations into the transcriptional regulation of chicken JUN can offer a molecular foundation for elucidating its mechanistic role in SSCs. Methods: In this study, we successfully cloned a 2000 bp upstream sequence of the JUN transcription start site and constructed a series of pGL3 recombinant vectors containing JUN promoters of varying lengths. Results: We verified the promoter activity of the 2000 bp upstream sequence by assessing the fluorescence intensity of DF-1 and identified the promoter activities of different regions via dual-luciferase assays. The transcription of JUN and its promoter region spanning -700 to 0 bp was modulated by an activator of the JNK signaling pathway. Bioinformatics analysis revealed that this -700 to 0 bp region was highly conserved among avian species and predicted the presence of binding sites for Wilms tumor 1 (WT1) and CCAAT/enhancer binding protein alpha (CEBPA). The JNK signaling pathway activator was found to upregulate the expression of these transcription factors in DF-1 cells. Through the deletion of binding sites and the overexpression of WT1 and CEBPA, we demonstrated that WT1 inhibited the transcription of JUN, while CEBPA promoted it. Conclusions: In conclusion, the -700 to 0 bp region is the key region of the JUN promoter, with WT1 inhibiting JUN transcription. The results of the study not only provide ideas for exploring the regulatory mechanism of JUN in chicken SSCs, but also lay an important foundation for the study of avian SSCs.
Collapse
Affiliation(s)
- Ruihong Kong
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jieyao Shi
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Ke Xie
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Han Wu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Xu Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Yani Zhang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009, China;
| | - Yingjie Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (R.K.); (J.S.); (K.X.); (H.W.); (X.W.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
3
|
Cozmin M, Lungu II, Gutu C, Stefanache A, Duceac LD, Șoltuzu BD, Damir D, Calin G, Bogdan Goroftei ER, Grierosu C, Boev M. Turmeric: from spice to cure. A review of the anti-cancer, radioprotective and anti-inflammatory effects of turmeric sourced compounds. Front Nutr 2024; 11:1399888. [PMID: 38863589 PMCID: PMC11165187 DOI: 10.3389/fnut.2024.1399888] [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: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 06/13/2024] Open
Abstract
Turmeric (Curcuma longa) has been extensively studied for its diverse pharmacological properties, including its potential role as an anticancer agent, antioxidant, and radioprotector. This review provides an overview of the chemical composition of turmeric, focusing on its main bioactive compounds, such as curcuminoids and volatile oils. Curcumin, the most abundant curcuminoid in turmeric, has been widely investigated for its various biological activities, including anti-inflammatory, antioxidant, and anticancer effects. Numerous in vitro and in vivo studies have demonstrated the ability of curcumin to modulate multiple signaling pathways involved in carcinogenesis, leading to inhibition of cancer cell proliferation, induction of apoptosis, and suppression of metastasis. Furthermore, curcumin has shown promising potential as a radioprotective agent by mitigating radiation-induced oxidative stress and DNA damage. Additionally, turmeric extracts containing curcuminoids have been reported to exhibit potent antioxidant activity, scavenging free radicals and protecting cells from oxidative damage. The multifaceted pharmacological properties of turmeric make it a promising candidate for the development of novel therapeutic strategies for cancer prevention and treatment, as well as for the management of oxidative stress-related disorders. However, further research is warranted to elucidate the underlying mechanisms of action and to evaluate the clinical efficacy and safety of turmeric and its bioactive constituents in cancer therapy and radioprotection. This review consolidates the most recent relevant data on turmeric's chemical composition and its therapeutic applications, providing a comprehensive overview of its potential in cancer prevention and treatment, as well as in radioprotection.
Collapse
Affiliation(s)
- Mihai Cozmin
- "Apollonia” University of Iasi, Faculty of Dental Medicine, Iași, Romania
| | | | - Cristian Gutu
- University Dunarea de Jos Faculty of Medicine and Pharmacy, Galați, Romania
- "Dr. Aristide Serfioti” Military Emergency Clinical Hospital, Galați, Romania
| | - Alina Stefanache
- "Grigore T. Popa" University of Medicine and Pharmacy, Iași, Romania
| | - Letitia Doina Duceac
- "Apollonia” University of Iasi, Faculty of Dental Medicine, Iași, Romania
- University Dunarea de Jos Faculty of Medicine and Pharmacy, Galați, Romania
- Prof. Dr. Nicolae Oblu” Neurosurg Hospital Iasi, 2 Ateneului, Iasi, Romania
| | | | - Daniela Damir
- "Grigore T. Popa" University of Medicine and Pharmacy, Iași, Romania
| | - Gabriela Calin
- "Apollonia” University of Iasi, Faculty of Dental Medicine, Iași, Romania
| | - Elena Roxana Bogdan Goroftei
- University Dunarea de Jos Faculty of Medicine and Pharmacy, Galați, Romania
- Sf. Ioan Emergency Clinical Hospital for Children, 2 Gheorghe Asachi Str., Galați, Romania
| | - Carmen Grierosu
- "Apollonia” University of Iasi, Faculty of Dental Medicine, Iași, Romania
| | - Monica Boev
- University Dunarea de Jos Faculty of Medicine and Pharmacy, Galați, Romania
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, Galați, Romania
| |
Collapse
|
4
|
Read CB, Ali AN, Stephenson DJ, Macknight HP, Maus KD, Cockburn CL, Kim M, Xie X, Carlyon JA, Chalfant CE. Ceramide-1-phosphate is a regulator of Golgi structure and is co-opted by the obligate intracellular bacterial pathogen Anaplasma phagocytophilum. mBio 2024; 15:e0029924. [PMID: 38415594 PMCID: PMC11005342 DOI: 10.1128/mbio.00299-24] [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: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Many intracellular pathogens structurally disrupt the Golgi apparatus as an evolutionarily conserved promicrobial strategy. Yet, the host factors and signaling processes involved are often poorly understood, particularly for Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis. We found that A. phagocytophilum elevated cellular levels of the bioactive sphingolipid, ceramide-1-phosphate (C1P), to promote Golgi fragmentation that enables bacterial proliferation, conversion from its non-infectious to infectious form, and productive infection. A. phagocytophilum poorly infected mice deficient in ceramide kinase, the Golgi-localized enzyme responsible for C1P biosynthesis. C1P regulated Golgi morphology via activation of a PKCα/Cdc42/JNK signaling axis that culminates in phosphorylation of Golgi structural proteins, GRASP55 and GRASP65. siRNA-mediated depletion of Cdc42 blocked A. phagocytophilum from altering Golgi morphology, which impaired anterograde trafficking of trans-Golgi vesicles into and maturation of the pathogen-occupied vacuole. Cells overexpressing phosphorylation-resistant versions of GRASP55 and GRASP65 presented with suppressed C1P- and A. phagocytophilum-induced Golgi fragmentation and poorly supported infection by the bacterium. By studying A. phagocytophilum, we identify C1P as a regulator of Golgi structure and a host factor that is relevant to disease progression associated with Golgi fragmentation.IMPORTANCECeramide-1-phosphate (C1P), a bioactive sphingolipid that regulates diverse processes vital to mammalian physiology, is linked to disease states such as cancer, inflammation, and wound healing. By studying the obligate intracellular bacterium Anaplasma phagocytophilum, we discovered that C1P is a major regulator of Golgi morphology. A. phagocytophilum elevated C1P levels to induce signaling events that promote Golgi fragmentation and increase vesicular traffic into the pathogen-occupied vacuole that the bacterium parasitizes. As several intracellular microbial pathogens destabilize the Golgi to drive their infection cycles and changes in Golgi morphology is also linked to cancer and neurodegenerative disorder progression, this study identifies C1P as a potential broad-spectrum therapeutic target for infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Curtis B. Read
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Anika N. Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Daniel J. Stephenson
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - H. Patrick Macknight
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth D. Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Chelsea L. Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, Florida, USA
| | - Xiujie Xie
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, USA
| | - Charles E. Chalfant
- Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, Virginia, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, Virginia, USA
| |
Collapse
|
5
|
Okonogi S, Chittasupho C, Sassa-deepaeng T, Khumpirapang N, Anuchpreeda S. Modification of Polyethylene Glycol-Hydroxypropyl Methacrylate Polymeric Micelles Loaded with Curcumin for Cellular Internalization and Cytotoxicity to Wilms Tumor 1-Expressing Myeloblastic Leukemia K562 Cells. Polymers (Basel) 2024; 16:917. [PMID: 38611175 PMCID: PMC11013463 DOI: 10.3390/polym16070917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Curcumin loaded in micelles of block copolymers of ω-methoxypoly(ethylene glycol) and N-(2-hydroxypropyl) methacrylamide modified with aliphatic dilactate (CD) or aromatic benzoyl group (CN) were previously reported to inhibit human ovarian carcinoma (OVCAR-3), human colorectal adenocarcinoma (Caco-2), and human lymphoblastic leukemia (Molt-4) cells. Myeloblastic leukemia cells (K562) are prone to drug resistance and differ in both cancer genotype and phenotype from the three mentioned cancer cells. In the present study, CD and CN micelles were prepared and their effects on K562 and normal cells were explored. The obtained CD and CN showed a narrow size distribution with diameters of 63 ± 3 and 50 ± 1 nm, respectively. The curcumin entrapment efficiency of CD and CN was similarly high, above 80% (84 ± 8% and 91 ± 3%). Both CD and CN showed suppression on WT1-expressing K562 and high cell-cycle arrest at the G2/M phase. However, CD showed significantly higher cytotoxicity to K562, with faster cellular uptake and internalization than CN. In addition, CD showed better compatibility with normal red blood cells and peripheral blood mononuclear cells than CN. The promising CD will be further investigated in rodents and possibly in clinical studies for leukemia treatment.
Collapse
Affiliation(s)
- Siriporn Okonogi
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Center of Excellent in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Center of Excellent in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tanongsak Sassa-deepaeng
- Agricultural Biochemistry Research Unit, Faculty of Sciences and Agricultural Technology, Rajamangala University of Technology Lanna Lampang, Lampang 52000, Thailand;
| | - Nattakanwadee Khumpirapang
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand;
| | - Songyot Anuchpreeda
- Center of Excellent in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
6
|
Wu J, Yan H, Xiang C. Wilms' tumor gene 1 in hematological malignancies: friend or foe? Hematology 2023; 28:2254557. [PMID: 37668240 DOI: 10.1080/16078454.2023.2254557] [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: 03/27/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023] Open
Abstract
Wilms' tumor gene 1 (WT1) is a transcription and post-translational factor that has a crucial role in the biological and pathological processes of several human malignancies. For hematological malignancies, WT1 overexpression or mutation has been found in leukemia and myelodysplastic syndrome. About 70-90% of acute myeloid leukemia patients showed WT1 overexpression, and 6-15% of patients carried WT1 mutations. WT1 has been widely regarded as a marker for monitoring minimal residual disease in acute myeloid leukemia. Many researchers were interested in developing WT1 targeting therapy. In this review, we summarized biological and pathological functions, correlation with other genes and clinical features, prognosis value and targeting therapy of WT1 in hematological features.
Collapse
Affiliation(s)
- Jie Wu
- Department of Emergency Medicine, The Fifth People's Hospital of Huai'an and Huai'an Hospital Affiliated to Yangzhou University, Huai'an, People's Republic of China
| | - Hui Yan
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, People's Republic of China
| | - Chunli Xiang
- Department of General Medicine, The Affiliated Huai'an Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, People's Republic of China
| |
Collapse
|
7
|
Rueankham L, Panyajai P, Saiai A, Rungrojsakul M, Tima S, Chiampanichayakul S, Yeerong K, Somwongin S, Chaiyana W, Dejkriengkraikul P, Okonogi S, Katekunlaphan T, Anuchapreeda S. Biological activities of extracts and compounds from Thai Kae-Lae (Maclura cochinchinensis (Lour.) Corner). BMC Complement Med Ther 2023; 23:191. [PMID: 37296375 DOI: 10.1186/s12906-023-03979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/27/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND AND AIMS The purpose of this study was to investigate the biological properties of Kae-Lae (Maclura cochinchinensis (Lour.) Corner), a traditional medicinal plant used in Ayurvedic recipes in Thailand. To achieve this objective, heartwood samples were collected from 12 sources across Thailand. Fractional extracts (n-hexane, ethyl acetate, and ethanol) and the dominant compounds (morin, resveratrol, and quercetin) were examined for their abilities on cytotoxicity, antioxidant, anti-inflammation, and antileukaemic activity (Wilms' tumour 1 protein was used as a well-known biomarker for leukaemic cell proliferation). METHODS The study used MTT to assess cytotoxicity in leukaemic cells (K562, EoL-1, and KG-1a). Antioxidant activities were evaluated using ABTS, DPPH, and FRAP assays. The anti-inflammatory activity was investigated by detecting IL-2, TNF-α, and NO using appropriate detection kits. Wilms' tumour 1 protein expression was measured by Western blotting to determine the anti-leukaemic activity. The inhibition of cell migration was also analyzed to confirm anticancer progression. RESULTS Among the tested extract fraction, ethyl acetate No. 001 displayed strong cytotoxicity specifically in EoL-1 cells, while n-hexane No. 008 demonstrated this effect in three cell lines. Resveratrol, on the other hand, displayed cytotoxicity in all the tested cells. Additionally, the three major compounds, morin, resveratrol, and quercetin, exhibited significant antioxidant and anti-inflammatory properties. In particular, resveratrol demonstrated a noteworthy decreased Wilms' tumour 1 protein expression and a reduction in cell proliferation across all cells. Moreover, ethyl acetate No. 001, morin, and resveratrol effectively inhibited MCF-7 cell migration. None of these compounds showed any impact on red blood cell haemolysis. CONCLUSION Based on these findings, it can be concluded that Kae-Lae has promising chemotherapeutic potential against leukaemic cells, with fractional extracts (ethyl acetate and n-hexane) and resveratrol exhibiting the most potent cytotoxic, antioxidant, anti-inflammatory, and anti-cell migration activities.
Collapse
Affiliation(s)
- Lapamas Rueankham
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pawaret Panyajai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Aroonchai Saiai
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Methee Rungrojsakul
- Department of Traditional Chinese Medicine, Faculty of Science, Chandrakasem, Rajabhat University, Bangkok, 10900, Thailand
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Kankanit Yeerong
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Suvimol Somwongin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wantida Chaiyana
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | - Siriporn Okonogi
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Trinnakorn Katekunlaphan
- Department of Chemistry, Faculty of Science, Chandrakasem, Rajabhat University, Bangkok, 10900, Thailand.
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
8
|
Luo J, Tugade T, Sun E, Pena Diaz AM, O’Gorman DB. Sustained AWT1 expression by Dupuytren's disease myofibroblasts promotes a proinflammatory milieu. J Cell Commun Signal 2022; 16:677-690. [PMID: 35414143 PMCID: PMC9733761 DOI: 10.1007/s12079-022-00677-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/22/2022] [Indexed: 12/13/2022] Open
Abstract
Palmar fibromatosis, also known as Dupuytren's disease (DD), is a common and heritable fibrosis of the hand. It is characterized by the formation of myofibroblastic nodules that can progress to palmar-digital contractures and permanent loss of dexterity. The presence of inflammatory cell infiltrate within these nodules has been interpreted to suggest a pathogenesis mediated by a proinflammatory microenvironment. However, the molecular mechanisms driving the formation of pro-fibrotic microenvironments in this and other fibroses remain unclear. To gain insights into this process, we have assessed the contributions of an alternatively spliced, multi-functional transcription factor, Wilms Tumor 1 (WT1), previously shown to be upregulated in primary myofibroblasts derived from DD tissues. Proinflammatory cytokine stimuli of DD myofibroblasts enhanced the expression of several distinct WT1 variants, the most sustained being a 5' truncated version of WT1, alternative WT1 (AWT1). Constitutive adenoviral expression of AWT1 in myofibroblasts derived from phenotypically non-fibrotic palmar fascia significantly induced the expression and secretion of proinflammatory cytokines, including some with potential as novel therapeutic targets. In summary, these data implicate roles for sustained AWT1 expression in DD as a transcriptional driver of a proinflammatory fascial milieu.
Collapse
Affiliation(s)
- Johnny Luo
- grid.39381.300000 0004 1936 8884Department of Biochemistry, University of Western Ontario, London, ON Canada
| | - Trisiah Tugade
- grid.39381.300000 0004 1936 8884Department of Biochemistry, University of Western Ontario, London, ON Canada
| | - Emmy Sun
- grid.39381.300000 0004 1936 8884Department of Biochemistry, University of Western Ontario, London, ON Canada
| | - Ana Maria Pena Diaz
- grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2 Canada
| | - David B. O’Gorman
- grid.39381.300000 0004 1936 8884Department of Biochemistry, University of Western Ontario, London, ON Canada ,grid.39381.300000 0004 1936 8884Department of Surgery, University of Western Ontario, London, ON Canada ,grid.415847.b0000 0001 0556 2414Lawson Health Research Institute, 268 Grosvenor Street, London, ON N6A 4V2 Canada
| |
Collapse
|
9
|
Antileukaemic Cell Proliferation and Cytotoxic Activity of Edible Golden Cordyceps ( Cordyceps militaris) Extracts. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5347718. [PMID: 35497915 PMCID: PMC9054435 DOI: 10.1155/2022/5347718] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022]
Abstract
Golden cordyceps (Cordyceps militaris) is a mushroom of the genus Cordyceps. It has been used as a food supplement for both healthy and ill people. In this study, the antileukaemic cell proliferation activities of golden cordyceps extracts were examined and compared with standard cordycepin (CDCP) in EoL-1, U937, and KG-1a cells. Wilms' tumour 1 (WT1) protein was used as a biomarker of leukaemic cell proliferation. The cytotoxicity of the extracts on leukaemic cells was determined using the MTT assay. Their inhibitory effects on WT1 protein expression and cell cycle progression of EoL-1 cells were investigated using Western blotting and flow cytometry, respectively. Induction of KG-1a cell differentiation (using CD11b as a marker) was determined using flow cytometry. The golden cordyceps extracts exhibited cytotoxic effects on leukaemic cells with the highest IC50 value of 16.5 ± 3.9 µg/mL, while there was no effect on normal blood cells. The expression levels of WT1 protein in EoL-1 cells were decreased after treatment with the extracts. Moreover, cell cycle progression and cell proliferation were inhibited. The levels of CD11b increased slightly following the treatment. All these findings confirm the antileukaemic proliferation activity of golden cordyceps.
Collapse
|
10
|
Wang S, Li Y, Li W, Zhang K, Yuan Z, Cai Y, Xu K, Zhou J, Du Z. Curcuma oil ameliorates benign prostatic hyperplasia through suppression of the nuclear factor-kappa B signaling pathway in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:113703. [PMID: 33340599 PMCID: PMC9586842 DOI: 10.1016/j.jep.2020.113703] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 11/20/2020] [Accepted: 11/27/2020] [Indexed: 05/05/2023]
Abstract
ETHNO PHARMACOLOGICAL RELEVANCE Curcuma longa L is traditionally used as an anti-inflammatory remedy in Chinese traditional medicine. Curcuma oil (CO), a lipophilic fraction from Curcuma longa L. has been reported to have anti-proliferative, anti-inflammatory and anti-oxidant activities. However, CO has never been investigated for its possible therapeutic effects on benign prostatic hyperplasia (BPH). AIMS OF THE STUDY The study is thus to determine the therapeutic effects of curcuma oil on BPH and also the possible mechanism (s) of action. MATERIALS &METHODS A BPH-1 cell line and Sprague Dawley (SD) rats were used to establish BPH models in vitro and in vivo, respectively. Rats were treated by CO (2.4, 7.2 mg/kg/i.g.) and finasteride (5 mg/kg/i.g.), respectively. Histological changes were examined by hematoxylin and eosin (H&E) staining. Protein expression was analyzed for 5α-reductase (5AR), dihydrotestosterone (DHT), interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α by ELISA. Ki-67, Caspase-8,-9 and -3 expressions were evaluated via immunohistochemistry (IHC). RESULTS CO effectively induced apoptosis in BPH-1 cells. BPH was successfully established by administration of testosterone propionate (TP) in rats, which upregulated both 5α-reductase expression and DHT production. Importantly, TP establishment significantly stimulated the phosphorylation of p65, one subunit of NF-κB, thus led to activation of the NF-κB signaling pathway in prostatic tissues of rats. In turn, the activation of NF-κB pathway induced concomitant upregulation of proinflammatory factors IL-1β, IL-6, TNF-α, and COX-2 and significant increase of the Bcl2/Bax expression ratio for enhanced cell survival, contributing to the initiation and progression of BPH in rats. Notably, CO therapy significantly decreased prostate weight and hyperplasia in BPH-induced animals. Also CO was found to suppress the expression of 5α-reductase and thus the production of DHT, which is essential for the amelioration of BPH. More importantly, CO was shown to suppress the activation of NF-κB pathway through decreasing the expression of phosphorylated p65 and consequently reduced the inflammatory responses and cell survival in prostatic tissues, leading to the inhibition of BPH development in rats. CONCLUSION Curcuma oil is very effective for ameliorating BPH in rats. The underlying mechanisms involve in reduced inflammatory responses and cell survival through suppression of the NF-κB signaling pathway by CO in prostatic tissues.
Collapse
Affiliation(s)
- Shanshan Wang
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Yun Li
- R&D Centre, Infinitus (China) Company Ltd, Guangzhou, China
| | - Wenzhi Li
- R&D Centre, Infinitus (China) Company Ltd, Guangzhou, China
| | - Kun Zhang
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Zhengqiang Yuan
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Yina Cai
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Kuncheng Xu
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China
| | - Jinrong Zhou
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.
| | - Zhiyun Du
- School of Biomedical and Phamaceutical Sciences, Gunagdong University of Technology, Guangzhou, 511400, China; Conney Allan Biotechnology Company Ltd, Guangzhou, 510095, China.
| |
Collapse
|
11
|
Gao W, Jin Z, Zheng Y, Xu Y. Psoralen inhibits the inflammatory response and mucus production in allergic rhinitis by inhibiting the activator protein 1 pathway and the downstream expression of cystatin‑SN. Mol Med Rep 2021; 24:652. [PMID: 34278468 PMCID: PMC8299190 DOI: 10.3892/mmr.2021.12291] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Psoralen (PSO) exerts anti-inflammatory pharmacological effects and plays an important role in a variety of inflammatory diseases. However, the effects of PSO with allergic rhinitis (AR) are yet to be reported. In the present study, an in vitro AR model was generated by inducing JME/CF15 human nasal epithelial cells with IL-13, after which MTT was used to assess the cytotoxicity of PSO. The expression levels of inflammatory cytokines (granulocyte-macrophage colony-stimulating factor and Eotaxin) were determined by ELISA. Furthermore, the expression of inflammatory IL-6 and −8, as well as mucin 5AC, was assessed by reverse transcription-quantitative PCR and western blotting, and cellular reactive oxygen species were detected using a 2′,7′-dichlorodihydrofluorescein diacetate fluorescent probe. Western blotting was also used to detect the expression and phosphorylation of c-Fos and c-Jun in the activator protein 1 (AP-1) pathway, as well as the expression of cystatin-SN (CST1). PSO inhibited the inflammatory response and mucus production in IL-13-induced JME/CF15 cells. Furthermore, the levels of c-Fos and c-Jun phosphorylation in the AP-1 pathway were decreased in IL-13-induced JME/CF15 cells following PSO treatment. The expression of pathway proteins was activated by the addition of PMA, an AP-1 pathway activator, which concurrently reversed the inhibitory effects of PSO on the inflammatory response and mucus formation. The addition of an AP-1 inhibitor (SP600125) further inhibited pathway activity, and IL-13-induced inflammation and mucus formation was restored. In conclusion, PSO regulates the expression of CST1 by inhibiting the AP-1 pathway, thus suppressing the IL-13-induced inflammatory response and mucus production in nasal mucosal epithelial cells.
Collapse
Affiliation(s)
- Wenying Gao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Zhenglong Jin
- Department of Neurology, Jiangmen Wuyi Hospital of TCM Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, Guangdong 529000, P.R. China
| | - Yanxia Zheng
- Department of TCM Pediatrics, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Youjia Xu
- Department of TCM Pediatrics, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
12
|
The regulatory role of SFRP5/WNT5A axis in allergic rhinitis through inhibiting JNK pathway activation and lowering mucin generation in human nasal epithelial cells. Exp Mol Pathol 2020; 118:104591. [PMID: 33285209 DOI: 10.1016/j.yexmp.2020.104591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 09/17/2020] [Accepted: 12/01/2020] [Indexed: 12/29/2022]
Abstract
Allergic rhinitis (AR) is tightly associated with type 2 inflammation. SFRP5 combined with WNT5A mainly inhibits chronic inflammatory response, atherosclerosis, and other metabolic disorders. However, the effect of SFRP5/WNT5A axis on recombinant human interleukin-13 (rhIL-13)-induced inflammation has not been studied. In this study, we aimed to investigate whether secreted frizzled-related protein 5 (SFRP5) could modulate the production of cytokines relevant to eosinophil infiltration and mucin secretion through blocking the activation of Wnt family 5A (WNT5A) signaling pathway. A mouse model of AR demonstrated low expression of SFRP5 and high expression of WNT5A, and indicated that the number of eosinophil and goblet cells was increased, concomitant with elevated IL-13, colony stimulating factor 2 (CSF2), chemokine ligand 11 (CCL11), Mucin 4, and Mucin 5AC levels. Furthermore, lentivirus-SFRP5 overexpression up-regulated the expression of SFRP5 but down-regulated WNT5A level, and inhibited the activation of JNK pathway via decreasing p-JNK1/2 (Thr183/Tyr185) and p-c-Jun (Ser73) protein expressions in rhIL-13-treated human nasal epithelial cells (HNEpCs). Noticeably, SFRP5 overexpression markedly reduced rhIL-13-induced inflammatory protein and mucin generation through lowered CSF2, CCL11, Mucin 4, as well as Mucin 5AC levels. Taken together, these findings confirmed the regulatory role of SFRP5/WNT5A axis in rhIL-13-mediated inflammatory response in HNEpCs.
Collapse
|
13
|
Cytotoxic and Antiproliferative Effects of Diarylheptanoids Isolated from Curcuma comosa Rhizomes on Leukaemic Cells. Molecules 2020; 25:molecules25225476. [PMID: 33238470 PMCID: PMC7700379 DOI: 10.3390/molecules25225476] [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] [Scholar Register] [Received: 10/21/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 12/31/2022] Open
Abstract
Curcuma comosa belongs to the Zingiberaceae family. In this study, two natural compounds were isolated from C. comosa, and their structures were determined using nuclear magnetic resonance. The isolated compounds were identified as 7-(3,4-dihydroxyphenyl)-5-hydroxy-1-phenyl-(1E)-1-heptene (1) and trans-1,7-diphenyl-5-hydroxy-1-heptene (2). Compound 1 showed the strongest cytotoxicity effect against HL-60 cells, while its antioxidant and anti-inflammatory properties were stronger than those of compound 2. Compound 1 proved to be a potent antioxidant, compared to ascorbic acid. Neither compounds had any effect on red blood cell haemolysis. Furthermore, compound 1 significantly decreased Wilms' tumour 1 protein expression and cell proliferation in KG-1a cells. Compound 1 decreased the WT1 protein levels in a time- and dose- dependent manner. Compound 1 suppressed cell cycle at the S phase. In conclusion, compound 1 has a promising chemotherapeutic potential against leukaemia.
Collapse
|
14
|
Mosaddad SA, Beigi K, Doroodizadeh T, Haghnegahdar M, Golfeshan F, Ranjbar R, Tebyanian H. Therapeutic applications of herbal/synthetic/bio-drug in oral cancer: An update. Eur J Pharmacol 2020; 890:173657. [PMID: 33096111 DOI: 10.1016/j.ejphar.2020.173657] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/01/2020] [Accepted: 10/20/2020] [Indexed: 12/18/2022]
Abstract
Oral cancer, as one of the most prevalent and invasive cancers that invade local tissue, can cause metastasis, and have high mortality. In 2018, around 355,000 worldwide oral cancers occurred and resulted in 177,000 deaths. Estimates for the year 2020 include about 53,260 new cases added to previous year's cases, and the estimated death toll from this cancer in 2020 is about 10,750 deaths more than previous years. Despite recent advances in cancer diagnosis and treatment, unfortunately, 50% of people with cancer cannot be cured. Of course, it should be remembered that the type of treatment used greatly influences patient recovery. There are not many choices when it comes to treating oral cancer. Research efforts focusing on the discovery and evolution of innovative therapeutic approaches for oral cancer are essential. Such traditional methods of treating this type of cancer like surgery and chemotherapy, have evolved dramatically during the past thirty to forty years, but they continue to cause panic among patients due to their side effects. Therefore, it is necessary to study and use drugs that are less risky for the patient as well as to provide solutions to reduce chemotherapy-induced adverse events that prevent many therapeutic risks. As mentioned above, this study examines low-risk therapies such as herbal remedies, biological drugs, and synthetic drugs in the hope that they will be useful to physicians, researchers, and scientists around the world.
Collapse
Affiliation(s)
- Seyed Ali Mosaddad
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kimia Beigi
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayebeh Doroodizadeh
- Department of Pediatric Dentistry, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maral Haghnegahdar
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Golfeshan
- Orthodontic Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Tebyanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
WT1 activates transcription of the splice factor kinase SRPK1 gene in PC3 and K562 cancer cells in the absence of corepressor BASP1. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2020; 1863:194642. [PMID: 33017668 DOI: 10.1016/j.bbagrm.2020.194642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022]
Abstract
Dysregulated alternative splicing plays a prominent role in all hallmarks of cancer. The splice factor kinase SRPK1 drives the activity of oncogenic splice factors such as SRSF1. SRSF1 in turn promotes the expression of splice isoforms that favour tumour growth, including proangiogenic VEGF. Knockdown (with siRNA) or chemical inhibition (using SPHINX) of SRPK1 in K562 leukemia and PC3 prostate cancer cell lines reduced cell proliferation, invasion and migration. In glomerular podocytes, the Wilms tumour suppressor zinc-finger transcription factor WT1 represses SRPK1 transcription. Here we show that in cancer cells WT1 activates SRPK1 transcription, unless a canonical WT1 binding site adjacent to the transcription start site is mutated. The ability of WT1 to activate SRPK1 transcription was reversed by the transcriptional corepressor BASP1, and both WT1 and BASP1 co-precipitated with the SRPK1 promoter. BASP1 significantly increased the expression of the antiangiogenic VEGF165b splice isoform. We propose that by upregulating SRPK1 transcription WT1 can direct an alternative splicing landscape that facilitates tumour growth.
Collapse
|
16
|
Anuchapreeda S, Chueahongthong F, Viriyaadhammaa N, Panyajai P, Anzawa R, Tima S, Ampasavate C, Saiai A, Rungrojsakul M, Usuki T, Okonogi S. Antileukemic Cell Proliferation of Active Compounds from Kaffir Lime ( Citrus hystrix) Leaves. Molecules 2020; 25:molecules25061300. [PMID: 32178481 PMCID: PMC7144100 DOI: 10.3390/molecules25061300] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 11/16/2022] Open
Abstract
Kaffir lime (Citrus hystrix) is a plant member of family Rutaceae, and its leaves are commonly used in folk medicine. The present study explores antileukemic effects of the extracts and purified active compounds from the leaves. The antileukemic activity was investigated via inhibition of Wilms’ tumor 1 (WT1), which is a protein that involves in leukemic cell proliferation. In addition, the compounds were investigated for their effects on WT1 gene expression using real time RT-PCR and Western blotting. Cell cycle arrest and total cell number were investigated using flow cytometry and trypan blue exclusion method, respectively. The results demonstrated that the hexane fractionated extract had the greatest inhibitory effect on WT1 gene expression of many leukemic cell lines and significantly decreased WT1 protein levels of K562 cells (representative of the leukemic cells), in a dose- and time-dependent manner. Subfraction No. 9 (F9) after partial purification of hexane fractioned extract showed the highest suppression on WT1 protein and suppressed cell cycle at G2/M. The organic compounds were isolated from F9 and identified as phytol and lupeol. The bioassays confirmed antiproliferative activities of natural products phytol and lupeol. The results demonstrated anticancer activity of the isolated phytol and lupeol to decrease leukemic cell proliferation.
Collapse
Affiliation(s)
- Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (F.C.); (N.V.); (P.P.); (S.T.)
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Tokyo 102-8554, Japan;
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: (S.A.); (T.U.); (S.O.); Tel.: +66-5394-9237 (S.A.); +81-3-3238-3446 (T.U.); +66-5394-1512 (S.O.)
| | - Fah Chueahongthong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (F.C.); (N.V.); (P.P.); (S.T.)
| | - Natsima Viriyaadhammaa
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (F.C.); (N.V.); (P.P.); (S.T.)
| | - Pawaret Panyajai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (F.C.); (N.V.); (P.P.); (S.T.)
| | - Riki Anzawa
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Tokyo 102-8554, Japan;
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (F.C.); (N.V.); (P.P.); (S.T.)
- Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Aroonchai Saiai
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Methee Rungrojsakul
- College of Alternative Medicine, Chandrakasem Rajabhat University, Bangkok 10900, Thailand;
| | - Toyonobu Usuki
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, Tokyo 102-8554, Japan;
- Correspondence: (S.A.); (T.U.); (S.O.); Tel.: +66-5394-9237 (S.A.); +81-3-3238-3446 (T.U.); +66-5394-1512 (S.O.)
| | - Siriporn Okonogi
- Research Center of Pharmaceutical Nanotechnology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
- Correspondence: (S.A.); (T.U.); (S.O.); Tel.: +66-5394-9237 (S.A.); +81-3-3238-3446 (T.U.); +66-5394-1512 (S.O.)
| |
Collapse
|
17
|
Saosathan S, Khounvong J, Rungrojsakul M, Katekunlaphan T, Tima S, Chiampanichayakul S, Berkland C, Anuchapreeda S. Costunolide and parthenolide from Champi Sirindhorn ( Magnolia sirindhorniae) inhibit leukemic cell proliferation in K562 and molt-4 cell lines. Nat Prod Res 2019; 35:988-992. [PMID: 31135208 DOI: 10.1080/14786419.2019.1610752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The magnolia plant has been used in traditional medicine since ancient times. This study was designed to investigate the effects of active compounds from Thai Champi Sirindhorn (Magnolia sirindhorniae) on leukemic biomarker Wilms' tumor 1 (WT1) protein expressions in K562 and Molt-4 cells. Costunolide (1) and parthenolide (2) were the major components used in this study, they were purified from ethyl acetate fractions. Costunolide (1) and parthenolide (2) had strong cytotoxic effects in K562 and Molt-4 cells measured with MTT assays. Their activities were compared to standard commercial costunolide (3) and parthenolide (4). Costunolide (1) and parthenolide (2) decreased WT1 protein levels and total cell numbers in K562 and Molt-4 cells. Both purified costunolide (1) and standard commercial costunolide (3) decreased WT1 protein levels in a time- and dose-dependent manner. Therefore, the active compounds from M. sirindhorniae were identified as promising sources for bioactive compounds for further applications in traditional medicine.
Collapse
Affiliation(s)
- Sawalee Saosathan
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Jalernsri Khounvong
- Department of Medical Sciences, Ministry of Public Health, Drug Section, Regional Medical Sciences Center 1 Chiang Mai, Chiang Mai, Thailand
| | - Methee Rungrojsakul
- College of Alternative Medicine, Chandrakasem Rajabhat University, Bangkok, Thailand
| | - Trinnakorn Katekunlaphan
- Department of Chemistry, Faculty of Science, Chandrakasem Rajabhat University, Bangkok, Thailand
| | - Singkome Tima
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand.,Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sawitree Chiampanichayakul
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Songyot Anuchapreeda
- Division of Clinical Microscopy, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai, Thailand.,Cancer Research Unit of Associated Medical Sciences (AMS CRU), Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|