1
|
Obrzut O, Gostyńska-Stawna A, Kustrzyńska K, Stawny M, Krajka-Kuźniak V. Curcumin: A Natural Warrior Against Inflammatory Liver Diseases. Nutrients 2025; 17:1373. [PMID: 40284236 PMCID: PMC12030243 DOI: 10.3390/nu17081373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Curcumin (CUR), a bioactive compound found in turmeric, has garnered attention for its potential anti-inflammatory properties and impact on liver health. Numerous studies suggest that CUR may be crucial in mitigating liver inflammation. The compound's anti-inflammatory effects are believed to be attributed to its ability to modulate various molecular pathways involved in the inflammatory response. Research indicates that CUR may suppress the activation of inflammatory cells and the production of pro-inflammatory cytokines in the liver. Additionally, it has been observed to inhibit the activity of transcription factors that play a key role in inflammation. By targeting these molecular mechanisms, CUR may help alleviate the inflammatory burden on the liver. Moreover, CUR's antioxidant properties are thought to contribute to its protective effects on the liver. Oxidative stress is closely linked to inflammation, and CUR's ability to neutralize free radicals may further support its anti-inflammatory action. While the evidence is promising, it is essential to note that more research is needed to fully understand the precise mechanisms through which CUR influences liver inflammation. Nevertheless, these findings suggest that CUR could be a potential therapeutic agent in managing liver inflammatory conditions. In this review, we explore the potential impact of CUR on inflammation, highlighting the key mechanisms involved, as reported in the literature.
Collapse
Affiliation(s)
- Olga Obrzut
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| | - Aleksandra Gostyńska-Stawna
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Karolina Kustrzyńska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, Bukowska 70, 60-812 Poznan, Poland
| | - Maciej Stawny
- Department of Pharmaceutical Chemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland; (A.G.-S.); (M.S.)
| | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznan, Poland;
| |
Collapse
|
2
|
Hernandez-Unzueta I, Telleria-Gonzalez U, Aransay AM, Martin Rodriguez JE, Sanz E, Márquez J. Unravelling the antitumor mechanism of Ocoxin through cancer cell genomics. Front Pharmacol 2025; 16:1540217. [PMID: 40176904 PMCID: PMC11961970 DOI: 10.3389/fphar.2025.1540217] [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: 12/05/2024] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Many therapies are being used to treat this disease, however, new treatments are now being implemented, since they are not always effective and their secondary effects represent one of the main reasons for cancer patients' loss of life quality during the progression of the disease. In this scenario, Ocoxin is a mixture of plant extracts, amino acids, vitamins and minerals, known for its antioxidant, anti-inflammatory and immunoregulatory properties, which has shown to exert antitumor effects in many cancers. The aim of this study is to elucidate the mechanism of action of the compound in colorectal cancer, triple negative breast cancer, pancreatic cancer and prostate cancer. Analyses performed through RNA sequencing revealed that the main effect of Ocoxin appears to be the alteration of cell metabolism, especially inducing the process of ferroptosis. Nevertheless, the modulation of the cell cycle was also remarkable. Ocoxin altered 13 genes in common in all the four cancers that were not only associated to metabolism and cell cycle but were also involved in the integrated stress response and unfolded protein response, suggesting that the compound causes the induction of cell death through several pathways. Although the mechanisms vary according to the type of cancer, this study highlights the potential of Ocoxin as an adjunctive treatment to improve outcomes in cancer therapy.
Collapse
Affiliation(s)
- Iera Hernandez-Unzueta
- Cell Biology and Histology Department, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
| | - Uxue Telleria-Gonzalez
- Cell Biology and Histology Department, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
| | - Ana María Aransay
- Genome Analysis Platform, CIC Biogune, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Joana Márquez
- Cell Biology and Histology Department, Faculty of Medicine and Nursing, University of the Basque Country, Leioa, Spain
| |
Collapse
|
3
|
Thongpon P, Intuyod K, Pongking T, Priprem A, Chomwong S, Tanasuka P, Mahalapbutr P, Suriya U, Vaeteewoottacharn K, Pinlaor P, Pinlaor S. Curcumin-Loaded Maltodextrin-Based Proniosomes Potentially Effective against Gemcitabine-Resistant Cholangiocarcinoma. ACS APPLIED BIO MATERIALS 2025; 8:913-930. [PMID: 39772434 PMCID: PMC11752495 DOI: 10.1021/acsabm.4c01832] [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: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
Cholangiocarcinoma (CCA) or bile-duct cancer is most prevalent in Southeast Asian counties including Thailand. Patients present at an advanced stage when the cancer is often drug resistant, leading to chemotherapy failure. Curcumin has therapeutic potential with various anticancer properties. However, its effectiveness is limited by its low bioavailability, poor solubility, and instability. This study aimed to synthesize, characterize and evaluate the efficacy of curcumin-loaded maltodextrin-based proniosomes (CMPNs) to overcome the limitations of curcumin for treating gemcitabine-resistant CCA cells (KKU-213BGemR) in vitro and in vivo. Various proniosome formulations were developed and tested for their efficacy against KKU-213BGemR cells using cytotoxicity, clonogenic, migration, and invasion assays. The potential mechanism involving in cell cycle arrest, apoptosis, expression of C/EBP homologous protein (CHOP), a pro-apoptotic transcription factor, and other apoptotic markers were investigated. The results showed that nanoscale CMPNs exhibited a good curcumin loading capacity and an entrapment efficiency of over 97%, as well as good stability and permeability through porcine esophageal mucosa. CMPNs inhibited proliferation, colony formation, migration/invasion and induced apoptosis in KKU-213BGemR cells. Western blot analysis revealed CMPNs significantly increased CHOP, the cleavage products of poly(ADP-ribose) polymerase-1 (PARP-1), apoptosis-inducing factor, and caspase-3 expression in KKU-213BGemR cells. A xenograft model revealed that 62.5 mg/kg BW CMPNs significantly suppressed proliferating cell nuclear antigen and increased CHOP-mediated apoptosis, leading to significantly reduced tumor volume. In conclusion, CMPNs effectively overcome limitations of curcumin and offer an effective strategy against gemcitabine-resistant CCA via CHOP-mediated pathways. These proniosomes are promising as an alternative treatment approach for CCA.
Collapse
Affiliation(s)
- Phonpilas Thongpon
- Department
of Parasitology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Kitti Intuyod
- Department
of Pathology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Thatsanapong Pongking
- Biomedical
Sciences Program, Graduate School, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Aroonsri Priprem
- Faculty
of Pharmacy, Mahasarakham University, Khamriang Sub-District, Kantarawichai
District, Mahasarakham 44150, Thailand
| | - Sasitorn Chomwong
- Department
of Parasitology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Pakornkiat Tanasuka
- Department
of Pathology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Panupong Mahalapbutr
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
| | - Utid Suriya
- Department
of Biochemistry, Faculty of Science, Mahidol
University, Bangkok 10400, Thailand
| | - Kulthida Vaeteewoottacharn
- Department
of Biochemistry, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Porntip Pinlaor
- Centre
for Research and Development in Medical Diagnostic Laboratory, Faculty
of Associated Medical Sciences, Khon Kaen
University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Somchai Pinlaor
- Department
of Parasitology, Faculty of Medicine, Khon
Kaen University, Khon Kaen 40002, Thailand
- Cholangiocarcinoma
Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
4
|
Liu B, Tian H, Momeni MR. The interplay of exercise and green tea: a new road in cancer therapy. Cancer Cell Int 2025; 25:6. [PMID: 39773739 PMCID: PMC11705833 DOI: 10.1186/s12935-024-03632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
Exercise is one of the most important activities for every individual due to its proven health beneficials. Several investigations have highlighted the advantageous impacts of aerobic exercise, largely attributed to its capacity to enhance the body's capability to defend against threats against oxidative stress. The information currently accessible suggests that adding regular aerobic exercise to a daily routine greatly decreases the chances of developing serious cancer and passing away. An unevenness in the levels of free radicals and the body's antioxidant defenses, made up of enzyme and non-enzyme antioxidants, results in oxidative pressure. Generally, an imbalance in the levels of oxidative stress triggers the creation of harmful reactive oxygen or nitrogen compounds, causing the development or progression of numerous ailments, including cancer. The equilibrium between pro-oxidant and antioxidant substances is a direct indicator of this imbalance. Green tea and its derivatives are rich sources of bioactive substances such as flavonoids and polyphenols which possess antioxidant abilities. Moreover, modulation of epigenetic targets as well as inflammatory pathways including ERK1/2 and NF-κB are other proposed mechanisms for its antioxidant activity. Recent studies demonstrate the promise of green tea as an antioxidant, showing its ability to decrease the likelihood of developing cancer by impacting actions like cell growth, blood vessel formation, and spread of cancer cells. This summary will concentrate on the complex network of different pathways related to physical activity and consumption of green tea. In particular, the focus of this research will be on examining how oxidative stress contributes to health and investigating the potential antioxidant properties of green tea, and the interconnected relationship between exercise and green tea in the treatment of cancer. Elucidation of these different pathways would help scientists for development of better therapeutic targets and further increase of current anticancer agents efficiency.
Collapse
Affiliation(s)
- Bing Liu
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Heyu Tian
- Henan University of Chinese Medicine, Zhengzhou, 450000, Henan, China.
| | | |
Collapse
|
5
|
Song W, Zhang Q, Cao Z, Jing G, Zhan T, Yuan Y, Kang N, Zhang Q. Targeting SERCA2 in Anti-Tumor Drug Discovery. Curr Drug Targets 2025; 26:1-16. [PMID: 39323343 DOI: 10.2174/0113894501325497240918042654] [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: 04/23/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
SERCA2, a P-type ATPase located on the endoplasmic reticulum of cells, plays an important role in maintaining calcium balance within cells by transporting calcium from the cytoplasm to the endoplasmic reticulum against its concentration gradient. A multitude of studies have demonstrated that the expression of SERCA2 is abnormal in a wide variety of tumor cells. Consequently, research exploring compounds that target SERCA2 may offer a promising avenue for the development of novel anti-tumor drugs. This review has summarized the anti-tumor compounds targeting SERCA2, including thapsigargin, dihydroartemisinin, curcumin, galangin, etc. These compounds interact with SERCA2 on the endoplasmic reticulum membrane, disrupting intracellular calcium ion homeostasis, leading to tumor cell apoptosis, autophagy and cell cycle arrest, ultimately producing anti-tumor effects. Additionally, several potential research directions for compounds targeting SERCA2 as clinical anti-cancer drugs have been proposed in the review. In summary, SERCA2 is a promising anti-tumor target for drug discovery and development.
Collapse
Affiliation(s)
- Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhiyong Cao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
6
|
Lucke-Wold B, Zasler ND, Ruchika FNU, Weisman S, Le D, Brunicardi J, Kong I, Ghumman H, Persad S, Mahan D, Delawan M, Shah S, Aghili-Mehrizi S. Supplement and nutraceutical therapy in traumatic brain injury. Nutr Neurosci 2024:1-35. [DOI: 10.1080/1028415x.2024.2404782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Affiliation(s)
| | - Nathan D. Zasler
- Founder, CEO & CMO, Concussion Care Centre of Virginia, Ltd., Medical Director, Tree of Life, Richmond, VA, USA
- Professor, affiliate, Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University, Richmond, VA, USA
- Professor, Visiting, Department of Physical Medicine and Rehabilitation, University of Virginia, Charlottesville, VA, USA
- Vice-Chairperson, IBIA, London, UK
- Chair Emeritus, IBIA, London, UK
| | - FNU Ruchika
- Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | - Sydney Weisman
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Dao Le
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Jade Brunicardi
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Iris Kong
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Haider Ghumman
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Sarah Persad
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - David Mahan
- Department of Internal Medicine, University of South Florida, Tampa, FL, USA
| | - Maliya Delawan
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | | |
Collapse
|
7
|
Zhang X, Han Y, Fan C, Jiang Y, Jiang W, Zheng C. Epigallocatechin gallate induces apoptosis in multiple myeloma cells through endoplasmic reticulum stress induction and cytoskeletal disruption. Int Immunopharmacol 2024; 141:112950. [PMID: 39159563 DOI: 10.1016/j.intimp.2024.112950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy that has prompted investigations into new potential therapeutic avenues. Epigallocatechin-3-gallate (EGCG), a major component of green tea, confers antioxidant, anti-inflammatory, and anti-tumor properties. Previous studies have shown that EGCG inhibits proliferation and induces apoptosis of multiple myeloma cells, however its underlying molecular mechanisms are largely unknown. In this study, we accordingly sought to examine the therapeutic effects and underlying mechanisms of EGCG on MM. Initially, using CCK8 (Cell Counting Kit-8) assays and Annexin V-FITC/PI staining, we demonstrated that EGCG dose-dependently reduced cell viability and induced apoptosis in the MM cell lines MM.1S and RPMI 8226. Subsequently, mRNA sequencing of EGCG-treated MM.1S cells revealed a significant upregulation of genes associated with endoplasmic reticulum stress (ERS), including P-eIF2α (phosphorylation-eukaryotic translation initiation factor 2 alpha), ATF4 (activating transcription factor 4), CHOP (C/EBP homologous protein, DDIT3), and PUMA (p53 upregulated modulator of apoptosis, BBC3), which were confirmed at the protein level by western blotting. Furthermore, treatment with the eIF2α inhibitor ISRIB reduced the rates of EGCG-induced apoptosis and promoted increases in the protein expression of all four ER stress-related molecules in MM cells. Additionally, mRNA-seq data revealed a downregulation of α-Tubulin 1b (TUBA1B) expression in EGCG-treated MM cells, which was confirmed by western blotting and immunofluorescence analyses. Moreover, we utilized a mouse model to show that EGCG inhibited myeloma tumor growth, which was inhibited by ISRIB. In summary, the findings of this novel study indicated that EGCG promotes apoptosis of MM cells, both via activation of the ER stress pathway and disruption of cytoskeletal integrity. These findings highlight the multi-faceted anti-tumor effects of EGCG and its potential clinical application in MM treatment.
Collapse
Affiliation(s)
- Xunqi Zhang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China; Shengli Oilfield Central Hospital, Dongying 257034, China
| | - Yanxiao Han
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China; Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China; Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China
| | - Chenliu Fan
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China; Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China; Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China
| | - Yang Jiang
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China; Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China; Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China.
| | - Wen Jiang
- Institute of Medical Science, The Second Hospital of Shandong University, Jinan, Shandong, China.
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital of Shandong University, Jinan, Shandong, China; Institute of Biotherapy for Hematological Malignancy, Shandong University, Jinan, Shandong, China; Shandong University-Karolinska Institute Collaboration Laboratory for Stem Cell Research, Jinan, Shandong, China.
| |
Collapse
|
8
|
Heydarzadeh S, Moshtaghie AA, Daneshpour M, Hedayati M. A novel approach to regulate glucose uptake in an anaplastic thyroid cancer cell line. Endocr Connect 2024; 14:EC-24-0336. [PMID: 39555605 PMCID: PMC11728915 DOI: 10.1530/ec-24-0336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 11/19/2024]
Abstract
AIMS AND BACKGROUND Curcumin's function in affecting cancer metabolic reprogramming remains poorly understood. Herein, we aimed to elucidate a novel link between Curcumin and the glucose uptake metabolism and glucose transporters (GLUTs) status in SW1736 cell line derived from anaplastic thyroid cancer. MATERIALS AND METHODS TheMTT test and flow cytometry was employed to test cell viability and cell death. For glucose uptake detection, ''GOD-PAP'' enzymatic colorimetric assay was applied to measure the direct glucose levels inside of the cells. Determination of GLUT1 and GLUT3 mRNA and protein expression in SW1736 cells was performed by qRT-PCR and western blotting. Also, the scratch wound healing assay was conducted for cell migration. RESULTS The data indicated that Curcumin-induced cell death is independent of apoptosis in this type of thyroid cancer cell line. Furthermore, significantly reduced GLUT1 and GLUT3 expression was observed after treatment with Curcumin, resulting in the inhibition of glucose uptake (p < 0.05). Scratch assay indicated the inhibition of cell migration in SW1736 cells treated by Curcumin (p < 0.05). CONCLUSION It can be concluded that GLUTs as metabolic targets can be blocked specifically by Curcumin for thyroid cancer prevention. Curcumin, as a promising anti-cancer agent, inhibits the growth of SW1736 anaplastic thyroid cancer cell line by regulating glucose uptake pathway and cell death. Altogether, these results suggest that the glucose pathway may be an important target for therapeutic intervention to sensitize tumor cells to cell death process by inhibition of glucose transporters.
Collapse
Affiliation(s)
- Shabnam Heydarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Asghar Moshtaghie
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Maryam Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Al Azzani M, Nizami ZN, Magramane R, Sekkal MN, Eid AH, Al Dhaheri Y, Iratni R. Phytochemical-mediated modulation of autophagy and endoplasmic reticulum stress as a cancer therapeutic approach. Phytother Res 2024; 38:4353-4385. [PMID: 38961675 DOI: 10.1002/ptr.8283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024]
Abstract
Autophagy and endoplasmic reticulum (ER) stress are conserved processes that generally promote survival, but can induce cell death when physiological thresholds are crossed. The pro-survival aspects of these processes are exploited by cancer cells for tumor development and progression. Therefore, anticancer drugs targeting autophagy or ER stress to induce cell death and/or block the pro-survival aspects are being investigated extensively. Consistently, several phytochemicals have been reported to exert their anticancer effects by modulating autophagy and/or ER stress. Various phytochemicals (e.g., celastrol, curcumin, emodin, resveratrol, among others) activate the unfolded protein response to induce ER stress-mediated apoptosis through different pathways. Similarly, various phytochemicals induce autophagy through different mechanisms (namely mechanistic target of Rapamycin [mTOR] inhibition). However, phytochemical-induced autophagy can function either as a cytoprotective mechanism or as programmed cell death type II. Interestingly, at times, the same phytochemical (e.g., 6-gingerol, emodin, shikonin, among others) can induce cytoprotective autophagy or programmed cell death type II depending on cellular contexts, such as cancer type. Although there is well-documented mechanistic interplay between autophagy and ER stress, only a one-way modulation was noted with some phytochemicals (carnosol, capsaicin, cryptotanshinone, guangsangon E, kaempferol, and δ-tocotrienol): ER stress-dependent autophagy. Plant extracts are sources of potent phytochemicals and while numerous phytochemicals have been investigated in preclinical and clinical studies, the search for novel phytochemicals with anticancer effects is ongoing from plant extracts used in traditional medicine (e.g., Origanum majorana). Nonetheless, the clinical translation of phytochemicals, a promising avenue for cancer therapeutics, is hindered by several limitations that need to be addressed in future studies.
Collapse
Affiliation(s)
- Mazoun Al Azzani
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rym Magramane
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed N Sekkal
- Department of Surgery, Specialty Orthopedic, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yusra Al Dhaheri
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
10
|
Wu MH, Hsieh YH, Lin CL, Ying TH, Hsia SM, Hsieh SC, Lee CH, Lin CL. Licochalcone A induces endoplasmic reticulum stress-mediated apoptosis of endometrial cancer cells via upregulation of GRP78 expression. ENVIRONMENTAL TOXICOLOGY 2024; 39:2961-2969. [PMID: 38308464 DOI: 10.1002/tox.24156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 02/04/2024]
Abstract
Licochalcone A (LicA), a natural compound extracted from licorice root, has been shown to exert a variety of anticancer activities. Whether LicA has such effects on endometrial cancer (EMC) is unclear. This study aims to investigate the antitumor effects of LicA on EMC. Our results show that LicA significantly reduced the viability and induced apoptosis of EMC cells and EMC-7 cells from EMC patients. LicA was also found to induce endoplasmic reticulum (ER) stress, leading to increased expression of ER-related proteins (GRP78/PERK/IRE1α/CHOP) in EMC cell lines. Suppression of GRP78 expression in human EMC cells treated with LicA significantly attenuated the effects of LicA, resulting in reduced ER-stress mediated cell apoptosis and decreased expression of ER- and apoptosis-related proteins. Our findings demonstrate that LicA induces apoptosis in EMC cells through the GRP78-mediated ER-stress pathway, emphasizing the potential of LicA as an anticancer therapy for EMC.
Collapse
Affiliation(s)
- Min-Hua Wu
- Laboratory Department, Chung-Kang Branch, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Min Hsia
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Shu-Ching Hsieh
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chien-Hsing Lee
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of China Medical University, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chu-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
11
|
Li S, Fan R, Wang Y, He K, Xu J, Li H. Application of calcium overload-based ion interference therapy in tumor treatment: strategies, outcomes, and prospects. Front Pharmacol 2024; 15:1352377. [PMID: 38425645 PMCID: PMC10902152 DOI: 10.3389/fphar.2024.1352377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
Low selectivity and tumor drug resistance are the main hinderances to conventional radiotherapy and chemotherapy against tumor. Ion interference therapy is an innovative anti-tumor strategy that has been recently reported to induce metabolic disorders and inhibit proliferation of tumor cells by reordering bioactive ions within the tumor cells. Calcium cation (Ca2+) are indispensable for all physiological activities of cells. In particular, calcium overload, characterized by the abnormal intracellular Ca2+ accumulation, causes irreversible cell death. Consequently, calcium overload-based ion interference therapy has the potential to overcome resistance to traditional tumor treatment strategies and holds promise for clinical application. In this review, we 1) Summed up the current strategies employed in this therapy; 2) Described the outcome of tumor cell death resulting from this therapy; 3) Discussed its potential application in synergistic therapy with immunotherapy.
Collapse
Affiliation(s)
- Shuangjiang Li
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Ruicheng Fan
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Yuekai Wang
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Kunqian He
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
- Battalion, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Jinhe Xu
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| | - Hongli Li
- Chongqing Key Laboratory of Neurobiology, Department of Teaching Experiment Center, College of Basic Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Hu Y, Cheng L, Du S, Wang K, Liu S. Antioxidant curcumin induces oxidative stress to kill tumor cells (Review). Oncol Lett 2024; 27:67. [PMID: 38192657 PMCID: PMC10773205 DOI: 10.3892/ol.2023.14200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
Curcumin is a plant polyphenol in turmeric root and a potent antioxidant. It binds to antioxidant response elements for gene regulation by nuclear factor erythroid 2-related factor 2, thereby suppressing reactive oxygen species (ROS) and exerting anti-inflammatory, anti-infective and other pharmacological effects. Of note, curcumin induces oxidative stress in tumors. It binds to several enzymes in tumors, such as carbonyl reductases, glutathione S-transferase P1 and nicotinamide adenine dinucleotide phosphate to induce mitochondrial damage, increase ROS production and ultimately induce tumor cell death. However, the instability and poor pharmacokinetic profile of curcumin in vivo limit its clinical application. Therefore, the effects of curcumin in vivo may be enhanced through its combination with drugs, derivative development and nanocarriers. In the present review, the mechanisms of curcumin that induce tumor cell death through oxidative stress are discussed. In addition, the methods used to enhance the antitumor activity of curcumin are described. Finally, the existing knowledge on the functions of curcumin in tumors, particularly in terms of oxidative stress, are summarized to facilitate future curcumin research.
Collapse
Affiliation(s)
- Ye Hu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Lei Cheng
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Shuguang Du
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Kesi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| | - Shuangping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, P.R. China
| |
Collapse
|
13
|
Hernández-Oliveras A, Zarain-Herzberg A. The role of Ca 2+-signaling in the regulation of epigenetic mechanisms. Cell Calcium 2024; 117:102836. [PMID: 37988873 DOI: 10.1016/j.ceca.2023.102836] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/23/2023]
Abstract
Epigenetic mechanisms regulate multiple cell functions like gene expression and chromatin conformation and stability, and its misregulation could lead to several diseases including cancer. Epigenetic drugs are currently under investigation in a broad range of diseases, but the cellular processes involved in regulating epigenetic mechanisms are not fully understood. Calcium (Ca2+) signaling regulates several cellular mechanisms such as proliferation, gene expression, and metabolism, among others. Moreover, Ca2+ signaling is also involved in diseases such as neurological disorders, cardiac, and cancer. Evidence indicates that Ca2+ signaling and epigenetics are involved in the same cellular functions, which suggests a possible interplay between both mechanisms. Ca2+-activated transcription factors regulate the recruitment of chromatin remodeling complexes into their target genes, and Ca2+-sensing proteins modulate their activity and intracellular localization. Thus, Ca2+ signaling is an important regulator of epigenetic mechanisms. Moreover, Ca2+ signaling activates epigenetic mechanisms that in turn regulate genes involved in Ca2+ signaling, suggesting possible feedback between both mechanisms. The understanding of how epigenetics are regulated could lead to developing better therapeutical approaches.
Collapse
Affiliation(s)
- Andrés Hernández-Oliveras
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Angel Zarain-Herzberg
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
14
|
Xu W, Shen Y. Curcumin affects apoptosis of colorectal cancer cells through ATF6-mediated endoplasmic reticulum stress. Chem Biol Drug Des 2024; 103:e14433. [PMID: 38230779 DOI: 10.1111/cbdd.14433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 01/18/2024]
Abstract
Colorectal cancer (CRC) is the main cause of cancer-associated death. Herein, we treated SW620 and HT-29 CRC cells with different curcumin concentrations, followed by treatment with the half maximal inhibitory concentration (IC50) curcumin/endoplasmic reticulum stress (ERS) inhibitor 4-phenyl butyric acid (4-PBA)/activating transcription factor 6 (ATF6) interference plasmid (si-ATF6). We detected cell proliferation/apoptosis, ATF6 cellular localization/nuclear translocation, ion concentration, ATF6 protein/apoptotic protein (Bax/Bcl-2/Cleaved Caspase-3) levels, and ERS-related proteins (glucose-regulated protein 78 [Grp78]/C/EBP homologous protein [CHOP]). We discovered inhibited cell proliferation/growth, enhanced cell apoptosis/(Bax/Bcl-2) ratio/Cleaved Caspase-3 levels/Ca2+ concentration in the cytoplasm/ERS-related protein (Grp78/CHOP) levels, and activated ERS following treatment with IC50 curcumin. 4-PBA partially reversed the inhibitory effect of curcumin on SW620 cells by restraining ERS. Curcumin stimulated ATF6 expression and its nuclear translocation to activate ERS. ATF6 silencing partly annulled the inhibitory effect of curcumin on SW620 cells. Our study explored the molecular mechanism of curcumin affecting CRC cell apoptosis through ATF6.
Collapse
Affiliation(s)
- Wei Xu
- Oncology Department of Integrated Traditional Chinese and Western Medicine, Hangzhou Cancer Hospital, Hangzhou, China
| | - Yu Shen
- Health Management Center, Hangzhou Wuyunshan Hospital (Hangzhou Institute of Health Promotion), Hangzhou, China
| |
Collapse
|
15
|
Ghaeini Hesarooeyeh Z, Basham A, Sheybani-Arani M, Abbaszadeh M, Salimi Asl A, Moghbeli M, Saburi E. Effect of resveratrol and curcumin and the potential synergism on hypertension: A mini-review of human and animal model studies. Phytother Res 2024; 38:42-58. [PMID: 37784212 DOI: 10.1002/ptr.8023] [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: 03/20/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
Resveratrol (RES) and curcumin (CUR) are two of the most extensively studied bioactive compounds in cardiovascular research from the past until today. These compounds have effectively lowered blood pressure by downregulating the renin-angiotensin system, exerting antioxidant effects, and exhibiting antiproliferative activities on blood vessels. This study aims to summarize the results of human and animal studies investigating the effects of CUR, RES, and their combination on hypertension and the molecular mechanisms involved. The published trials' results are controversial regarding blood pressure reduction with different doses of RES and CUR, highlighting the need to address this issue.
Collapse
Affiliation(s)
- Zahra Ghaeini Hesarooeyeh
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ayoub Basham
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Mahshid Abbaszadeh
- Student Research Committee, School of Dentistry, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Girigoswami K, Pallavi P, Girigoswami A. Intricate subcellular journey of nanoparticles to the enigmatic domains of endoplasmic reticulum. Drug Deliv 2023; 30:2284684. [PMID: 37990530 PMCID: PMC10987057 DOI: 10.1080/10717544.2023.2284684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023] Open
Abstract
It is evident that site-specific systemic drug delivery can reduce side effects, systemic toxicity, and minimal dosage requirements predominantly by delivering drugs to particular pathological sites, cells, and even subcellular structures. The endoplasmic reticulum (ER) and associated cell organelles play a vital role in several essential cellular functions and activities, such as the synthesis of lipids, steroids, membrane-associated proteins along with intracellular transport, signaling of Ca2+, and specific response to stress. Therefore, the dysfunction of ER is correlated with numerous diseases where cancer, neurodegenerative disorders, diabetes mellitus, hepatic disorder, etc., are very common. To achieve satisfactory therapeutic results in certain diseases, it is essential to engineer delivery systems that can effectively enter the cells and target ER. Nanoparticles are highly biocompatible, contain a variety of cargos or payloads, and can be modified in a pliable manner to achieve therapeutic effectiveness at the subcellular level when delivered to specific organelles. Passive targeting drug delivery vehicles, or active targeting drug delivery systems, reduce the nonselective accumulation of drugs while reducing side effects by modifying them with small molecular compounds, antibodies, polypeptides, or isolated bio-membranes. The targeting of ER and closely associated organelles in cells using nanoparticles, however, is still unsymmetrically understood. Therefore, here we summarized the pathophysiological prospect of ER stress, involvement of ER and mitochondrial response, disease related to ER dysfunctions, essential therapeutics, and nanoenabled modulation of their delivery to optimize therapy.
Collapse
Affiliation(s)
- Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Pragya Pallavi
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, TN, India
| |
Collapse
|
17
|
Zhang L, Qiu L, Xu S, Cheng X, Wu J, Wang Y, Gao W, Bao J, Yu H. Curcumin induces mitophagy by promoting mitochondrial succinate dehydrogenase activity and sensitizes human papillary thyroid carcinoma BCPAP cells to radioiodine treatment. Toxicol In Vitro 2023; 93:105669. [PMID: 37634662 DOI: 10.1016/j.tiv.2023.105669] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/14/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Thyroid cancer is one of the most common endocrine malignancies. Differentiated thyroid cancer (DTC) treatment is based on the ability of thyroid follicular cells to accumulate radioactive iodide (RAI). DTC generally has a good prognosis. However, tumor dedifferentiation or defect in certain cell death mechanism occurs in a subset of DTC patients, leading to RAI resistance. Therefore, developing novel therapeutic approaches that enhance RAI sensitivity are still warranted. We found that curcumin, an active ingredient in turmeric with anti-cancer properties, rapidly accumulated in the mitochondria of thyroid cancer cells but not normal epithelial cells. Curcumin treatment triggered mitochondrial membrane depolarization, engulfment of mitochondria within autophagosomes and a robust decrease in mitochondrial mass and proteins, indicating that curcumin selectively induced mitophagy in thyroid cancer cells. In addition, curcumin-induced mitophagic cell death and its synergistic cytotoxic effect with radioiodine could be attenuated by autophagy inhibitor, 3-methyladenine (3-MA). Interestingly, the mechanism of mitophagy-inducing potential of curcumin was its unique mitochondria-targeting property, which induced a burst of SDH activity and excessive ROS production. Our data suggest that curcumin induces mitochondrial dysfunction and triggers lethal mitophagy, which synergizes with radioiodine to kill thyroid cancer cells.
Collapse
Affiliation(s)
- Li Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; School of Life science and Technology, Southeast University, Nanjing 210096, China.
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Shichen Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Xian Cheng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Jing Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Yunping Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenjing Gao
- School of Life science and Technology, Southeast University, Nanjing 210096, China
| | - Jiandong Bao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, Jiangsu, China
| |
Collapse
|
18
|
Manica D, Silva GBD, Silva APD, Marafon F, Maciel SFVDO, Bagatini MD, Moreno M. Curcumin promotes apoptosis of human melanoma cells by caspase 3. Cell Biochem Funct 2023; 41:1295-1304. [PMID: 37792322 DOI: 10.1002/cbf.3863] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/10/2023] [Accepted: 09/17/2023] [Indexed: 10/05/2023]
Abstract
Cutaneous melanoma (CM) is a malignant neoplasm with a high metastatic rate that shows poor response to systemic treatments in patients with advanced stages. Recently, studies have highlighted the antineoplastic potential of natural compounds, such as polyphenols, in the adjuvant therapy context to treat CM. The objective of the present study was to evaluate the effect of different concentrations of curcumin (0.1-100 µM) on the metastatic CM cell line SK-MEL-28. The cells were treated for 6 and 24 h with different concentrations of curcumin. Cell viability was assessed by 3-(4,5-dimethyl-2thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and fluorescence microscopy. The apoptotic-inducing potential was detected by annexin V flow cytometry. The wound healing assay was used to verify cell migration after the curcumin exposition. The redox profile was evaluated by levels of the pro-oxidant markers reactive oxygen species (ROS) and Nitric oxide (NOx) and antioxidants of total thiols (PSH) and nonprotein thiols. The gene expression and enzymatic activity of caspase 3 were evaluated by reverse transcription-quantitative polymerase chain reaction and a sensitive fluorescence assay, respectively. Curcumin significantly decreased the cell viability of SK-MEL-28 cells at both exposure times. It also induced apoptosis at the highest concentration tested (p < .0001). SK-MEL-28 cell migration was inhibited by curcumin after treatment with 10 µM (p < .0001) and 100 µM (p < .0001) for 6 and 24 h (p = .0006 and p < .0001, respectively). Furthermore, curcumin significantly increased levels of ROS and NOx. Finally, curcumin was capable of increasing the gene expression at 10 µM (p = .0344) and 100 µM (p = .0067) and enzymatic activity at 10 µM (p = .0086) and 100 µM (p < .0001) of caspase 3 after 24 h. For the first time, we elucidated in our study that curcumin increases ROS levels, promoting oxidative stress that activates the caspase pathway and culminates in SK-MEL-28 metastatic CM cell death.
Collapse
Affiliation(s)
- Daiane Manica
- Postgraduate Programme in Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Gilnei Bruno da Silva
- Multicentric Postgraduate Programme in Biochemistry and Molecular Biology, State University of Santa Catarina, Lages, Santa Catarina, Brazil
| | - Alana Patrícia da Silva
- Postgraduate Programme in Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Filomena Marafon
- Postgraduate Programme in Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Margarete Dulce Bagatini
- Postgraduate Programme in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, Santa Catarina, Brazil
| | - Marcelo Moreno
- Postgraduate Programme in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, Santa Catarina, Brazil
| |
Collapse
|
19
|
Silva Barcelos EC, Rompietti C, Adamo FM, Dorillo E, De Falco F, Del Papa B, Baldoni S, Nogarotto M, Esposito A, Capoccia S, Geraci C, Sorcini D, Stella A, Arcaleni R, Tini V, Imbroisi Valle Errera F, Rosati E, Sportoletti P. NOTCH1-mutated chronic lymphocytic leukemia displays high endoplasmic reticulum stress response with druggable potential. Front Oncol 2023; 13:1218989. [PMID: 37817771 PMCID: PMC10561002 DOI: 10.3389/fonc.2023.1218989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023] Open
Abstract
Introduction Constitutive activation of NOTCH1-wild-type (NT1-WT) signaling is associated with poor outcomes in chronic lymphocytic leukemia (CLL), and NOTCH1 mutation (c.7541_7542delCT), which potentiates NOTCH1 signaling, worsens the prognosis. However, the specific mechanisms of NOTCH1 deregulation are still poorly understood. Accumulative evidence mentioned endoplasmic reticulum (ER) stress/unfolded protein response (UPR) as a key targetable pathway in CLL. In this study, we investigated the impact of NOTCH1 deregulation on CLL cell response to ER stress induction, with the aim of identifying new therapeutic opportunities for CLL. Methods We performed a bioinformatics analysis of NOTCH1-mutated (NT1-M) and NT1-WT CLL to identify differentially expressed genes (DEGs) using the rank product test. Quantitative real-time polymerase chain reaction (qPCR), Western blotting, cytosolic Ca2+, and annexin V/propidium iodide (PI) assay were used to detect curcumin ER stress induction effects. A median-effect equation was used for drug combination tests. The experimental mouse model Eμ-TCL1 was used to evaluate the impact of ER stress exacerbation by curcumin treatment on the progression of leukemic cells and NOTCH1 signaling. Results and discussion Bioinformatics analysis revealed gene enrichment of the components of the ER stress/UPR pathway in NT1-M compared to those in NT1-WT CLL. Ectopic expression of NOTCH1 mutation upregulated the levels of ER stress response markers in the PGA1 CLL cell line. Primary NT1-M CLL was more sensitive to curcumin as documented by a significant perturbation in Ca2+ homeostasis and higher expression of ER stress/UPR markers compared to NT1-WT cells. It was also accompanied by a significantly higher apoptotic response mediated by C/EBP homologous protein (CHOP) expression, caspase 4 cleavage, and downregulation of NOTCH1 signaling in NT1-M CLL cells. Curcumin potentiated the apoptotic effect of venetoclax in NT1-M CLL cells. In Eμ-TCL1 leukemic mice, the administration of curcumin activated ER stress in splenic B cells ex vivo and significantly reduced the percentage of CD19+/CD5+ cells infiltrating the spleen, liver, and bone marrow (BM). These cellular effects were associated with reduced NOTCH1 activity in leukemic cells and resulted in prolonged survival of curcumin-treated mice. Overall, our results indicate that ER stress induction in NT1-M CLL might represent a new therapeutic opportunity for these high-risk CLL patients and improve the therapeutic effect of drugs currently used in CLL.
Collapse
Affiliation(s)
- Estevão Carlos Silva Barcelos
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo, Vitória, Brazil
| | - Chiara Rompietti
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Francesco Maria Adamo
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Erica Dorillo
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Filomena De Falco
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Stefano Baldoni
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Manuel Nogarotto
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Angela Esposito
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Silvia Capoccia
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Clelia Geraci
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Daniele Sorcini
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Arianna Stella
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Roberta Arcaleni
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | - Valentina Tini
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| | | | - Emanuela Rosati
- Department of Medicine and Surgery, Biosciences and Medical Embryology Section, University of Perugia, Perugia, Italy
| | - Paolo Sportoletti
- Department of Medicine and Surgery, Institute of Hematology, Centro di Ricerca Emato-Oncologica (CREO), University of Perugia, Perugia, Italy
| |
Collapse
|
20
|
任 丽, 邹 明, 朱 行, 徐 文, 刘 刚, 孙 俊, 范 方, 张 从. [Curcumin suppresses proliferation, migration and invasion of papillary thyriod cancer B-CPAP cells through the Keap1-Nrf2 pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1356-1362. [PMID: 37712272 PMCID: PMC10505584 DOI: 10.12122/j.issn.1673-4254.2023.08.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE To observe the effects of curcumin on migration and invasion of papillary thyriod cancer B-CPAP cells. METHODS B-CPAP cells were treated with 5, 10, 15, or 20 μmol/L curcumin, and the changes in cell survival, migration and invasion were examined using MTT assay and Transwell assay. ROS levels in the treated cells were detected with a DCFH-DA probe. The expression levels of Nrf2 and Keap1 in the cells were determined using Western blotting and qRT-PCR. RESULTS Treatment with curcumin dose- and time-dependently suppressed the viability of B-CPAP cells (P < 0.05 or P < 0.01). Curcumin inhibited the migration and invasion (P < 0.001) and promoted ROS production in B-CPAP cells in a dose-dependent manner, and application of NAC effectively reversed curcumin- induced increase of ROS. Curcumin at 20 μmol/L significantly decreased the protein and mRNA expressions of Nrf2 and increased the expressions of Keap1 protein and mRNA (P < 0.05 or P < 0.01), causing also significantly reduced expression of Nrf2 protein in the cell nuclei (P < 0.05) without obviously affecting its expression in the cytoplasm (P > 0.05). CONCLUSION Curcumin inhibits the proliferation, migration and invasion of papillary thyriod cancer B-CPAP cells probably via the Keap1-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- 丽 任
- 蚌埠医学院检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 明远 邹
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 行春 朱
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
- 蚌埠医学院癌症转化医学安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Cancer Translational Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 文隽 徐
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
- 蚌埠医学院癌症转化医学安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Cancer Translational Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 刚 刘
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 俊杰 孙
- 蚌埠医学院检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 方田 范
- 蚌埠医学院药学院//安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy/Anhui Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu Medical College, Bengbu 233030, China
| | - 从利 张
- 第一附属医院麻醉科,安徽 蚌埠 233004Department of Anesthesia, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
21
|
Sazonova EN, Gusev IA, Filatova TS. Effects of Non-Opiate Analogue of Leu-Enkephalin on the Ion Currents, Number of Nucleoli, and p53 Expression in Isolated Cardiomyocytes of Albino Rats. Bull Exp Biol Med 2023; 175:544-548. [PMID: 37768450 DOI: 10.1007/s10517-023-05902-2] [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: 02/27/2023] [Indexed: 09/29/2023]
Abstract
Acute exposure of isolated ventricular cardiomyocytes to non-opiate analogue of leu-enkephalin (NALE peptide: Phe-D-Ala-Gly-Phe-Leu-Arg) in a concentration of 100 μg/liter and 6-h incubation in NALE solution did not significantly change ATP-dependent K+ current, L-type Ca2+ current, p53 protein expression, and number of nucleoli in the cardiomyocyte nuclei. Incubation of cardiomyocytes with NALE (100 μg/liter) in combination with NOP receptor blocker J-113397 (1 mg/liter) was followed by an increase in Ca2+ L-type current and the number of p53+ cells. The exposure of cardiomyocytes to NALE in a concentration 1000 μg/liter induced similar changes in the studied parameters (increase in Ca2+ L-type current and number of p53+ cardiomyocytes); an increase in the mean number of nucleoli was also observed. Our findings suggest that NALE peptide has direct effect on cardiomyocytes and NOP receptors are involved in this effect.
Collapse
Affiliation(s)
- E N Sazonova
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia
- Khabarovsk Branch of Far-Eastern Research Center of Physiology and Pathology of Breath, Research Institute of Maternity and Childhood Protection, Khabarovsk, Russia
| | - I A Gusev
- Far Eastern State Medical University, Ministry of Health of the Russian Federation, Khabarovsk, Russia.
| | - T S Filatova
- Department of Human and Animal Physiology, Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
22
|
Jo SL, Yang H, Lee HW, Hong EJ. Curcumae radix Reduces Endoplasmic Reticulum Stress in Mice with Chronic Neuroinflammation. Biomedicines 2023; 11:2107. [PMID: 37626603 PMCID: PMC10452873 DOI: 10.3390/biomedicines11082107] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 08/27/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is a condition in which the ER protein-folding machinery is impaired, leading to the accumulation of improperly folded proteins and triggering an unfolded-protein response. Excessive ER stress causes cell death and contributes to the development of chronic diseases. Interestingly, there is a bidirectional relationship between ER stress and the nuclear factor-kappa B (NF-κB) pathway. Curcumin, a natural polyphenolic compound found in Curcumae radix, exerts its neuroprotective effects by regulating ER stress and inflammation. Therefore, investigating the potential protective and regulatory effects of curcumin on ER stress, inflammation, and neurodegeneration under chronic neuroinflammatory conditions is of great interest. Mice were pretreated with Curcumae radix extract (CRE) for 19 days and then treated with CRE plus lipopolysaccharide for 1 week. We monitored pro-inflammatory cytokine levels in the serum and ER stress-, inflammation-, and neurodegeneration-related markers in the mouse cerebrum and hippocampus using Western blotting and qRT-PCR. CRE reduced Interleukin-1 beta levels in the blood and brain of mice with lipopolysaccharide-induced chronic inflammation. CRE also suppressed the expression of markers related to the ER stress and NF-κB signaling pathways. The expression of neurodegeneration-related markers was reduced in the mouse cerebrum and hippocampus. CRE exerts neuroprotective effects under chronic inflammatory conditions via multifaceted anti-inflammatory and ER stress-pathway regulatory mechanisms.
Collapse
Affiliation(s)
- Seong-Lae Jo
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Hyun Yang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea;
| | - Hye Won Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea;
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
23
|
Yang M, Liu C, Jiang N, Liu Y, Luo S, Li C, Zhao H, Han Y, Chen W, Li L, Xiao L, Sun L. Endoplasmic reticulum homeostasis: a potential target for diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1182848. [PMID: 37383398 PMCID: PMC10296190 DOI: 10.3389/fendo.2023.1182848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/31/2023] [Indexed: 06/30/2023] Open
Abstract
The endoplasmic reticulum (ER) is the most vigorous organelle in intracellular metabolism and is involved in physiological processes such as protein and lipid synthesis and calcium ion transport. Recently, the abnormal function of the ER has also been reported to be involved in the progression of kidney disease, especially in diabetic nephropathy (DN). Here, we reviewed the function of the ER and summarized the regulation of homeostasis through the UPR and ER-phagy. Then, we also reviewed the role of abnormal ER homeostasis in residential renal cells in DN. Finally, some ER stress activators and inhibitors were also summarized, and the possibility of maintaining ER homeostasis as a potential therapeutic target for DN was discussed.
Collapse
Affiliation(s)
- Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chongbin Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
24
|
Hu C, Yan L, Li P, Yu Y. Identification of calcium metabolism related score associated with the poor outcome in papillary thyroid carcinoma. Front Oncol 2023; 13:1108773. [PMID: 37056339 PMCID: PMC10086330 DOI: 10.3389/fonc.2023.1108773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
IntroductionPapillary thyroid carcinoma is a type of thyroid cancer that exhibits significant variability in prognosis. Extensive research indicates that the impaired signaling of 1,25(OH)2D3-VDR may be a crucial factor in the development and progression of PTC.MethodsTo investigate this further, Integrated analysis mRNA expression information from The Cancer Genome Atlas and GEO, we compared gene expression in cancer and normal tissues and identified differentially expressed genes (DEGs). Through this analysis, we identified DEGs and calculated risk estimates for seven genetic markers.ResultsSubsequently, we constructed predictive models using LASSO-Cox regression to test the predictive value of these markers. Our results revealed that 64 calcium metabolism-related genes showed significant differences between tumor and normal tissues. Ten of the identified DEGs were significantly associated with overall survival, indicating their potential role in disease progression. Using the average risk score for the seven genetic markers, we divided patients into high- and low-risk groups. We found that patients in the low-risk group had significantly better overall survival than those in the high-risk group, highlighting the importance of these genetic markers in predicting prognosis. Further analysis using Cox regression demonstrated that the risk levels had independent predictive power. Additionally, we conducted functional analysis of the identified genetic markers, which showed significant differences in immune status between the two patient groups. We also investigated the effect of these calcium metabolism-related genes on thyroid cancer biological functions, immune microenvironment, and drug resistance.DiscussionOur findings provide evidence of a novel genetic signature associated with calcium metabolism, which can predict prognosis in patients with PTC. These results may have significant implications for the development of new diagnostic and therapeutic approaches to improve outcomes for PTC patients.
Collapse
Affiliation(s)
- Chuanxiang Hu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lijuan Yan
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Protein Sciences, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Peng Li
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Protein Sciences, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
- *Correspondence: Peng Li, ; Yang Yu,
| | - Yang Yu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- *Correspondence: Peng Li, ; Yang Yu,
| |
Collapse
|
25
|
Ozisik H, Ozdil B, Suner A, Sipahi M, Erdogan M, Cetinkalp S, Ozgen G, Saygili F, Oktay G, Aktug H. The expression of HDAC9 and P300 in papillary thyroid carcinoma cell line. Pathol Res Pract 2023; 243:154385. [PMID: 36857949 DOI: 10.1016/j.prp.2023.154385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Papillary thyroid carcinoma (PTC) is the most common type of thyroid cancer and accounts for 85-90% of all thyroid cancers. Metastatic differentiated thyroid cancer, radioiodine-refractory thyroid cancer, and anaplastic thyroid cancer still lack effective therapeutic options. Here, we aimed to assess HDAC9 and P300 expression in the papillary thyroid carcinoma cell line and compare them with normal thyroid cells. METHODS Nthy-ori-3-1, a normal thyroid cell line, and BCPAP, a PTC cell line, were cultured for 24 and 48 h and immunofluorescence staining was used to determine the levels of HDAC9 and P300 protein expression. HDAC9 paracrine release was assessed using an ELISA assay. RESULTS HDAC9 protein expression was higher in both cell groups at the 48th hour than at the 24th hour; however, P300 protein expression was lower in BCPAP cells at the 48th hour than at the 24th hour. In comparison to Nthy-ori-3-1, BCPAP expressed more HDAC9 and P300 proteins. HDAC9 secretion slightly increased in Nthy-ori-3-1 cells from 24 to 48 h. Furthermore, HDAC9 secretion in BCPAP cells dramatically decreased from 24 to 48 h. CONCLUSION Our findings revealed that the expression of HDAC9 and P300 was higher in the PTC cell line than in normal thyroid cells. This indicates that the acetylation mechanism in thyroid cancer cells is not the same as it is in healthy cells. Epigenetic studies may reveal the mechanisms underlying PTC with further analysis.
Collapse
Affiliation(s)
- Hatice Ozisik
- Ege University, Department of Endocrinology and Metabolism, İzmir, Turkey.
| | - Berrin Ozdil
- Ege University, Department of Histology and Embryology, İzmir, Turkey; Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Aslı Suner
- Ege University, Department of Biostatistics and Medical Informatics, İzmir, Turkey
| | - Murat Sipahi
- Dokuz Eylül University, Institue of Health Sciences, Department of Biochemistry, İzmir, Turkey
| | - Mehmet Erdogan
- Ege University, Department of Endocrinology and Metabolism, İzmir, Turkey
| | - Sevki Cetinkalp
- Ege University, Department of Endocrinology and Metabolism, İzmir, Turkey
| | - Gokhan Ozgen
- Ege University, Department of Endocrinology and Metabolism, İzmir, Turkey
| | - Fusun Saygili
- Ege University, Department of Endocrinology and Metabolism, İzmir, Turkey
| | - Gulgun Oktay
- Dokuz Eylül University, Department of Medical Biochemistry, İzmir, Turkey
| | - Huseyin Aktug
- Ege University, Department of Histology and Embryology, İzmir, Turkey
| |
Collapse
|
26
|
Rahimi A, Sharifi H, Li PCH. Cytosolic Calcium Measurement Utilizing a Single-Cell Biochip to Study the Effect of Curcumin and Resveratrol on a Single Glioma Cell. Methods Mol Biol 2023; 2689:13-25. [PMID: 37430043 DOI: 10.1007/978-1-0716-3323-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
A microfluidic method has been developed for real-time measurement of the effects of curcumin on the intracellular calcium concentration in a single glioma cell (U87-MG). This method is based on quantitative fluorescence measurement of intracellular calcium in a cell selected in a single-cell biochip. This biochip consists of three reservoirs, three channels, and a V-shaped cell retention structure. Because of the adherent nature of glioma cells, a single cell can adhere within the aforementioned V-shaped structure. The single-cell calcium measurement will minimize cell damage caused by conventional cell calcium assay methods. Previous studies have shown that curcumin increased cytosolic calcium in glioma cells using the fluorescent dye: Fluo-4. So in this study, the effects of 5 μM and 10 μM solutions of curcumin on the increases of cytosolic calcium in a single glioma cell have been measured. Moreover, the effects of 100 μM and 200 μM of resveratrol are measured. At the final stage of the experiments, ionomycin was used to increase the intracellular calcium to the highest possible level due to dye saturation. It has been demonstrated that microfluidic cell calcium measurement is a real-time cytosolic assay that requires small quantities of reagent, which will have potential uses for drug discovery.
Collapse
Affiliation(s)
- Abolfazl Rahimi
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Hamide Sharifi
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Paul C H Li
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada.
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
27
|
β-carotene alleviates LPS-induced inflammation through regulating STIM1/ORAI1 expression in bovine mammary epithelial cells. Int Immunopharmacol 2022; 113:109377. [DOI: 10.1016/j.intimp.2022.109377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
|
28
|
Wang S, Gao X, Li J, Wei S, Shao Y, Yin Y, Zhang D, Tang M. The anticancer effects of curcumin and clinical research progress on its effects on esophageal cancer. Front Pharmacol 2022; 13:1058070. [DOI: 10.3389/fphar.2022.1058070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
Esophageal cancer (EC) is a common tumor of the gastrointestinal system and a major threat to human health. The etiology and incidence of EC vary depending on the type of pathology. Owing to the unique physiological structure of the esophagus and the poor biological behavior of EC, the treatment modalities available are limited, and the prognosis of patients is relatively poor. Curcumin is a type of natural phytochemical belonging to the class of phenolic compounds. It exerts favorable anticancer effects on various cancers. A growing body of evidence indicates that curcumin suppresses tumor development and progression by inhibiting tumor cell proliferation, invasion, and migration, thus inducing apoptosis, regulating microRNA expression, reversing multidrug resistance, and inducing sensitivity to the therapeutic effect of chemoradiotherapy. Multiple cellular molecules, growth factors, and genes encoding proteins participating in different signaling pathways interact with each other to contribute to the complex and orderly anticancer effect. The efficacy and safety of curcumin have been established in preclinical studies for EC and clinical trials for other cancers. However, the low bioavailability of curcumin limits its clinical application. Therefore, the modification of curcumin analogs, the combination of curcumin with other drugs or therapies, and the use of novel nanocarriers have been widely investigated to improve the clinical effects of curcumin in EC.
Collapse
|
29
|
Liang Y, Huang Y, Shao R, Xiao F, Lin F, Dai H, Pan L. Propofol produces neurotoxicity by inducing mitochondrial apoptosis. Exp Ther Med 2022; 24:630. [PMID: 36160898 PMCID: PMC9468839 DOI: 10.3892/etm.2022.11567] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Propofol is a fast and short-acting intravenous anesthetic widely used in clinical anesthesia and intensive care unit sedation. However, its use can cause abnormal effects on the central nervous system. Thus, the purpose of this study was to investigate the mechanism of propofol on primary hippocampal neuron injury. In addition, we aimed to determine whether a correlation exists between propofol and mitochondrial apoptosis-induced neurotoxicity. Hippocampal neurons cultured for 4 days were exposed to different drugs. The treatment groups were divided according to drug exposure into propofol, a rotenone inhibitor, and a coenzyme Q10 agonist groups. The final concentrations of propofol were 1, 10 and 100 µM. The content of ATP and reactive oxygen species (ROS) in the neurons of each group were detected using commercial kits in the culture supernatant after 3 h of drug exposure. Western blotting was used to analyze the expression of apoptosis-related proteins. The JC-1 kit was used to detect the mitochondrial membrane potential. The results revealed that, compared with the non-propofol treatment groups, the expression of apoptosis-related proteins, ATP content, and mitochondrial membrane potential were significantly decreased while the ROS content was markedly increased in the propofol treatment group. In conclusion, propofol treatment promoted damage to hippocampal neuronal mitochondria in a dose-dependent manner. This damage may lead to neuronal apoptosis and neurotoxicity by inducing the inhibition of mitochondrial respiratory chain complex I.
Collapse
Affiliation(s)
- Yubing Liang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yu Huang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rongge Shao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
30
|
Curcumin Increased the Sensitivity of Non-Small-Cell Lung Cancer to Cisplatin through the Endoplasmic Reticulum Stress Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6886366. [PMID: 35754693 PMCID: PMC9232348 DOI: 10.1155/2022/6886366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 04/10/2022] [Accepted: 05/23/2022] [Indexed: 01/05/2023]
Abstract
Objective Non-small-cell lung cancer (NSCLC) is one of the most lethal cancers. Although cisplatin-based chemotherapies have been regarded as a promising treatment approach, cisplatin resistance still remains one of the major clinical challenges. Curcumin, a naturally occurring polyphenol, has been proved to increase chemotherapeutic efficiency of NSCLC cells. However, the role of curcumin in cisplatin-resistant NSCLC cells has been rarely investigated. This study aims to investigate whether curcumin enhances cisplatin sensitivity of human NSCLC cells and its underlying mechanisms. Method A549/DDP and H1299/DDP cells were treated by DDP or/and curcumin before cell viability, and apoptosis were determined by using a CCK-8 assay and flow cytometer. The expressions of apoptosis and ER stress-related proteins, including cleaved caspase-3, cleaved PARP, CHOP, GRP78, XBP-1, ATF6, and caspase-4, were measured by the qPCR and western blotting. After cotreatment by DDP and curcumin, A549/DDP and H1299/DDP cells were further treated by the ER stress inhibitor, salubrinal (20 μm), after which the cell apoptosis and viability were detected. Result Treatment by DDP and curcumin can substantially decrease cell viability, while can increase the cell apoptosis rate, elevate mRNA and protein expressions of apoptosis and ER stress-related proteins, compared with cells treated by DDP or curcumin alone. Salubrinal treatment can counteract the suppressive effect of DDP and curcumin on cell viability and decrease the cell apoptosis of A549/DDP and H1299/DDP cells. Conclusion Curcumin can increase the sensitivity of NSCLC to cisplatin through an ER stress pathway and thus can be served as one of the molecular targets for overcoming the cisplatin resistance.
Collapse
|
31
|
Zhang X, Jiang X, Yu A, Duan H. Aconitine induces brain tissue damage by increasing the permeability of the cerebral blood-brain barrier and over-activating endoplasmic reticulum stress. Am J Transl Res 2022; 14:3216-3224. [PMID: 35702066 PMCID: PMC9185023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 03/30/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE This study aimed to explore the neurotoxicity of aconitine and its underlying mechanism. METHODS The rats were administered with the aconitine solution intragastrically at different dosages (0.5 mg/kg, 1.5 mg/kg, and 2.5 mg/kg). Evans blue (EB) extravasation and evaluation of tight junction protein expression were performed to determine the permeability of the blood-brain barrier. Cellular damage, apoptosis, and levels of endoplasmic reticulum (ER) stress markers were determined using H&E staining, Tunnel assay, and western blotting. The effects of aconitine on cell viability, apoptosis, and activation of the ER stress signaling in PC12 cells were assessed in vitro using the MTT assay, flow cytometry, western blot, and immunofluorescence analysis. RESULTS Aconitine was observed to significantly increase the murine blood-brain barrier penetrability in a dose-dependent manner. The in vivo experimental results revealed that aconitine could stimulate the pathway for endoplasmic reticulum stress. The increase in the endoplasmic reticulum stress in the brain tissue promoted apoptosis, leading to brain damage. Moreover, PC12 cell proliferation was inhibited upon treatment with aconitine in a dose-dependent manner. In addition, cell apoptosis was increased upon treatment with aconitine also in a dose-dependent manner. These findings indicated that aconitine caused damage to PC12 cells via endoplasmic reticulum stress. CONCLUSION Aconitine induces brain tissue damage by increasing the penetrability of the blood-brain barrier in the cerebral region and over-activating the endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University Zunyi, Guizhou, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University Zunyi, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University Zunyi, Guizhou, China
| | - Haizhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University Zunyi, Guizhou, China
| |
Collapse
|
32
|
Curcumin Inhibits Papillary Thyroid Cancer Cell Proliferation by Regulating lncRNA LINC00691. Anal Cell Pathol 2022; 2022:5946670. [PMID: 35256924 PMCID: PMC8898135 DOI: 10.1155/2022/5946670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 02/07/2022] [Accepted: 02/13/2022] [Indexed: 12/19/2022] Open
Abstract
Papillary thyroid cancer (PTC) is a type of epithelial-derived differentiated TC that reportedly accounts for a majority of TCs. Curcumin, a polyphenolic compound and a member of the Zingiberaceae (ginger) family derived from turmeric plants, can exhibit anticancer effects. Herein, we aimed to investigate the effect of curcumin on PTC and elucidate underlying mechanisms. Accordingly, PTC B-CPAP cells were treated with curcumin, in combination with/without long noncoding RNA LINC00691 inhibition, to determine the effect of curcumin and its relationship with LINC00691 in PTC cells. We observed that curcumin treatment decreased B-CPAP cell proliferation and promoted apoptosis. Curcumin inhibited LINC00691 expression in B-CPAP cells. Curcumin administration or si-LINC00691 transfection alone promoted ATP levels, inhibited glucose uptake and lactic acid levels, and inhibited lactate dehydrogenase A and hexokinase 2 protein expression in B-CPAP cells, which were further enhanced by combination treatment. Moreover, curcumin administration or si-LINC00691 transfection alone inhibited p-Akt activity, further suppressed by combination treatment. Akt inhibition promoted apoptosis and suppressed the Warburg effect in B-CPAP cells. In conclusion, our findings indicate that curcumin promotes apoptosis and suppresses proliferation and the Warburg effect by inhibiting LINC00691 in B-CPAP cells. The precise molecular mechanism might be mediated through the Akt signaling pathway, providing a theoretical basis for the treatment of PTC with curcumin.
Collapse
|
33
|
Wang Y, Wang L, Luo R, Sun Y, Zou M, Wang T, Guo Q, Peng X. Glycyrrhizic Acid against Mycoplasma gallisepticum-Induced Inflammation and Apoptosis Through Suppressing the MAPK Pathway in Chickens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1996-2009. [PMID: 35128924 DOI: 10.1021/acs.jafc.1c07848] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mycoplasma gallisepticum (MG) is the primary pathogen of chronic respiratory diseases (CRDs) in chickens. In poultry production, antibiotics are mostly used to prevent and control MG infection, but the drug resistance and residue problems caused by them cannot be ignored. Glycyrrhizic acid (GA) is derived from licorice, a herb traditionally used to treat various respiratory diseases. Our study results showed that GA significantly inhibited the mRNA and protein expression of pMGA1.2 and GapA in vitro and in vivo. Furthermore, the network pharmacology study revealed that GA most probably resisted MG infection through the MAPK signaling pathway. Our results demonstrated that GA inhibited MG-induced expression of MMP2/MMP9 and inflammatory factors through the p38 and JUN signaling pathways, but not the ERK pathway in vitro. Besides, histopathological sections showed that GA treatment obviously attenuated tracheal and lung damage caused by MG invasion. In conclusion, GA can inhibit MG-triggered inflammation and apoptosis by suppressing the expression of MMP2/MMP9 through the JNK and p38 pathways and inhibit the expression of virulence genes to resist MG. Our results suggest that GA might serve as one of the antibiotic alternatives to prevent MG infection.
Collapse
Affiliation(s)
- Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Lulu Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Ronglong Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Yingfei Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Qiao Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| |
Collapse
|
34
|
Shakeri F, Bibak B, Safdari MR, Keshavarzi Z, Jamialahmadi T, Sathyapalan T, Sahebkar A. Cellular and molecular mechanisms of curcumin on thyroid gland disorders. Curr Med Chem 2022; 29:2878-2890. [PMID: 35142266 DOI: 10.2174/0929867329666220210145033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/13/2021] [Accepted: 11/21/2021] [Indexed: 11/22/2022]
Abstract
There is growing literature on the positive therapeutic potentials of curcumin. Curcumin or diferuloylmethane is a polyphenol obtained from the plant Curcuma longa. Curcumin has been used widely in Ayurvedic and Chinese medicine for various conditions. The role of curcumin on thyroid glands has been shown by its effects on various biological pathways, including anti-inflammatory, antioxidant, anti-proliferative, apoptosis, angiogenesis, cell cycle and metastasis. We reviewed the recent literature on curcumin applications for thyroid dysfunction, including hyperthyroidism and hypothyroidism, and discussed the molecular mechanisms of these effects. This review aims to summarize the wealth of research related to the thyroid gland therapeutic effect of curcumin.
Collapse
Affiliation(s)
- Farzaneh Shakeri
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Reza Safdari
- Department of Orthopedic Surgery, Imam Ali Hospital, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zakieh Keshavarzi
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Hong Q, Li X, Lin Q, Shen Z, Feng J, Hu C. Resveratrol Improves Intestinal Morphology and Anti-Oxidation Ability in Deoxynivalenol-Challenged Piglets. Animals (Basel) 2022; 12:ani12030311. [PMID: 35158635 PMCID: PMC8833336 DOI: 10.3390/ani12030311] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Deoxynivalenol (DON)-contaminated feed may cause anorexia, vomiting, immunosuppression, and intestinal dysfunction in pigs, which would lead to growth retardation and great losses in the pig industry. In this study, the effects of resveratrol (RES) on growth performance, the intestinal barrier, antioxidant capacity, and mitochondrial function in weaned pigs fed with DON-contaminated diets were investigated. Dietary supplementation with resveratrol increased the average daily feed intake of piglets. Diets supplemented with resveratrol increased the villus height and the ratio of the jejunum villus height to crypt depth, increased the activities of superoxide dismutase (SOD), and increased the total antioxidant capacity in the jejunum mucosa. After being supplemented with RES, the level of reactive oxygen species (ROS) in mitochondria was decreased, while the mitochondrial membrane potential in the jejunum was increased. In conclusion, these results suggested that resveratrol effectively relieved DON-induced oxidative stress in weaned piglets, improved intestinal barrier function, enhanced mitochondrial function, and improved the growth performance of piglets. Abstract This study aimed to investigate the potential effects of resveratrol (RES) on intestinal function and oxidative stress in deoxynivalenol (DON)-challenged piglets. Twenty-four healthy Duroc × Yorkshire × Landrace weaned piglets at the age of 28 ± 1 days were randomly divided into four groups with six repetitions per group. The four groups were as follows: the control group (CON), fed with a basic diet; the RES group, fed with a basal diet + 300 mg/kg RES; the DON group, fed with a basal diet containing 2.65 mg/kg DON; and the DON + RES group, fed with a basal diet containing 2.65 mg/kg DON + 300 mg/kg RES. The results showed that the growth performance and intestinal function of DON-challenged piglets were significantly decreased (p < 0.05). Compared with the DON group, the average daily feed intake of piglets in the DON + RES group was significantly increased (p < 0.05). Additionally, dietary RES ameliorated DON-induced intestinal morphology impairment, as indicated by the increased (p < 0.05) jejunal villi height and the ratio of the jejunal villi height/crypt depth. Furthermore, after the addition of RES, the activities of superoxide dismutase (SOD) and total antioxidant capacity (T-AOC) in the jejunum mucosa were significantly increased, and the content of malondialdehyde (MDA) was significantly declined (p < 0.05). In addition, the level of reactive oxygen species (ROS) in the mitochondria was significantly reduced by RES, while the mitochondrial membrane potential in jejunum was significantly increased by RES (p < 0.05). However, there was no obvious difference between DON + RES and DON groups on average daily gain and the ratio of feed togain, except for the significant inhibition of average daily feed intake (p < 0.05). In conclusion, RES could effectively alleviate the DON-induced oxidative stress on weaned piglets, and reduce the damage to mitochondria and intestinal morphology, so as to improve the growth performance of piglets.
Collapse
Affiliation(s)
- Qihua Hong
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
- Correspondence: (Q.H.); (C.H.)
| | - Xin Li
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Qian Lin
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Zhuojun Shen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Jie Feng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
| | - Caihong Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; (X.L.); (Q.L.); (Z.S.); (J.F.)
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou 310058, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou 310058, China
- Correspondence: (Q.H.); (C.H.)
| |
Collapse
|
36
|
Li X, Miao S, Li F, Ye F, Yue G, Lu R, Shen H, Ye Y. Cellular Calcium Signals in Cancer Chemoprevention and Chemotherapy by Phytochemicals. Nutr Cancer 2022; 74:2671-2685. [PMID: 35876249 DOI: 10.1080/01635581.2021.2020305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Xue Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Shuhan Miao
- Department of Health Care, Zhenjiang Fourth Peoples Hospital, Zhenjiang, China
| | - Feng Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Fen Ye
- Department of Clinical Laboratory Center, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Guang Yue
- Department of Internal Medicine, The Third Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
- Center for Experimental Research, Affiliated Kunshan Hospital, Jiangsu University, Kunshan, Suzhou, China
| | - Haijun Shen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
37
|
Maleki Dana P, Sadoughi F, Asemi Z, Yousefi B. The role of polyphenols in overcoming cancer drug resistance: a comprehensive review. Cell Mol Biol Lett 2022; 27:1. [PMID: 34979906 PMCID: PMC8903685 DOI: 10.1186/s11658-021-00301-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapeutic drugs are used to treat advanced stages of cancer or following surgery. However, cancers often develop resistance against drugs, leading to failure of treatment and recurrence of the disease. Polyphenols are a family of organic compounds with more than 10,000 members which have a three-membered flavan ring system in common. These natural compounds are known for their beneficial properties, such as free radical scavenging, decreasing oxidative stress, and modulating inflammation. Herein, we discuss the role of polyphenols (mainly curcumin, resveratrol, and epigallocatechin gallate [EGCG]) in different aspects of cancer drug resistance. Increasing drug uptake by tumor cells, decreasing drug metabolism by enzymes (e.g. cytochromes and glutathione-S-transferases), and reducing drug efflux are some of the mechanisms by which polyphenols increase the sensitivity of cancer cells to chemotherapeutic agents. Polyphenols also affect other targets for overcoming chemoresistance in cancer cells, including cell death (i.e. autophagy and apoptosis), EMT, ROS, DNA repair processes, cancer stem cells, and epigenetics (e.g. miRNAs).
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
38
|
Wang Y, Zhang X, Wen Y, Li S, Lu X, Xu R, Li C. Endoplasmic Reticulum-Mitochondria Contacts: A Potential Therapy Target for Cardiovascular Remodeling-Associated Diseases. Front Cell Dev Biol 2021; 9:774989. [PMID: 34858991 PMCID: PMC8631538 DOI: 10.3389/fcell.2021.774989] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular remodeling occurs in cardiomyocytes, collagen meshes, and vascular beds in the progress of cardiac insufficiency caused by a variety of cardiac diseases such as chronic ischemic heart disease, chronic overload heart disease, myocarditis, and myocardial infarction. The morphological changes that occur as a result of remodeling are the critical pathological basis for the occurrence and development of serious diseases and also determine morbidity and mortality. Therefore, the inhibition of remodeling is an important approach to prevent and treat heart failure and other related diseases. The endoplasmic reticulum (ER) and mitochondria are tightly linked by ER-mitochondria contacts (ERMCs). ERMCs play a vital role in different signaling pathways and provide a satisfactory structural platform for the ER and mitochondria to interact and maintain the normal function of cells, mainly by involving various cellular life processes such as lipid metabolism, calcium homeostasis, mitochondrial function, ER stress, and autophagy. Studies have shown that abnormal ERMCs may promote the occurrence and development of remodeling and participate in the formation of a variety of cardiovascular remodeling-associated diseases. This review focuses on the structure and function of the ERMCs, and the potential mechanism of ERMCs involved in cardiovascular remodeling, indicating that ERMCs may be a potential target for new therapeutic strategies against cardiovascular remodeling-induced diseases.
Collapse
Affiliation(s)
- Yu Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinrong Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ya Wen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Sixuan Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaohui Lu
- Emergency Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ran Xu
- Jinan Tianqiao People's Hospital, Jinan, China
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
39
|
Sharifi Noghabi H, Ahmed AQ, Li PCH. Intracellular Calcium Increases Due to Curcumin Measured Using a Single-Cell Biochip. ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1888967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Hamideh Sharifi Noghabi
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Chemistry, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Abdul Q. Ahmed
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Paul C. H. Li
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
40
|
New Insights into Curcumin- and Resveratrol-Mediated Anti-Cancer Effects. Pharmaceuticals (Basel) 2021; 14:ph14111068. [PMID: 34832850 PMCID: PMC8622305 DOI: 10.3390/ph14111068] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
Curcumin and resveratrol are bioactive natural compounds displaying anti-inflammatory, anti-oxidant and anti-cancer properties. In this study, we compared the cytotoxic effects of these molecules and the molecular mechanisms involved against Her-2/neu-positive breast and salivary cancer cell lines. We found that both curcumin and resveratrol were efficient in reducing cancer cell survival and that they differently affected autophagy, ROS and activation of the PI3K/AKT/mTOR pathway. Moreover, we found that resveratrol and curcumin in combination exerted a stronger cytotoxic effect in correlation with the induction of a stronger ER stress and the upregulation of pro-death UPR molecule CHOP. This effect also correlated with the induction of pro-survival autophagy by curcumin and its inhibition by resveratrol. In conclusion, this study unveils new molecular mechanisms underlying the anti-cancer effects of resveratrol, curcumin and their combination, which can help to design new therapeutic strategies based on the use of these polyphenols.
Collapse
|
41
|
Zhang L, Xu S, Cheng X, Wu J, Wu L, Wang Y, Wang X, Bao J, Yu H. Curcumin induces autophagic cell death in human thyroid cancer cells. Toxicol In Vitro 2021; 78:105254. [PMID: 34634291 DOI: 10.1016/j.tiv.2021.105254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/27/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022]
Abstract
Curcumin, a polyphenolic compound, is a well-known anticancer agent, although its poor bioavailability remains a big concern. Recent studies suggest that autophagy-targeted therapy may be a useful adjunct treatment for patients with thyroid cancer. Curcumin acts as an autophagy inducer on many cancer cells. However, little is known about the exact role of curcumin on thyroid cancer cells. In the present study, curcumin significantly inhibited the growth of thyroid cancer cells. Autophagy was markedly induced by curcumin treatment as evidenced by an increase in LC3-II conversion, beclin-1 accumulation, p62 degradation as well as the increased formation of acidic vesicular organelles (AVOs). 3-MA, an autophagy inhibitor, partially rescued thyroid cancer cells from curcumin-induced cell death. Additionally, curcumin was found to exert selective cytotoxicity on thyroid cancer cells but not normal epithelial cells and acted as an autophagy inducer through activation of MAPK while inhibition of mTOR pathways. Hyperactivation of the AKT/mTOR axis was observed in the majority of PTC samples we tested, and thyroid cancer cell lines along with cancer tissue specimens sustained a low basal autophagic activity. Taken together, our results provide new evidence that inducing autophagic cell death may serve as a potential anti-cancer strategy to handle thyroid cancer.
Collapse
Affiliation(s)
- Li Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; School of Life science and Technology, Southeast University, Nanjing 210096, China.
| | - Shichen Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Xian Cheng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Jing Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Liying Wu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Yunping Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaowen Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jiandong Bao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| |
Collapse
|
42
|
Preparation of Soluble Complex of Curcumin for the Potential Antagonistic Effects on Human Colorectal Adenocarcinoma Cells. Pharmaceuticals (Basel) 2021; 14:ph14090939. [PMID: 34577638 PMCID: PMC8467777 DOI: 10.3390/ph14090939] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/15/2021] [Accepted: 09/15/2021] [Indexed: 12/29/2022] Open
Abstract
This study was designed to investigate the effects of curcumin (CMN) soluble complex (SC) prepared by melt casting (HM) and hot-melt extrusion (HME) technology. Phase solubility (PS) study, in silico molecular modeling, aqueous solubility, drug release, and physicochemical investigation including a novel dyeing test was performed to obtain an optimized complex by a central composite design (CCD). The results show that the HME-SC produces better improvements towards solubility (0.852 ± 0.02), dissolution (91.87 ± 0.21% at 30 min), with an ideal stability constant (309 and 377 M−1 at 25 and 37 °C, respectively) and exhibits AL type of isotherm indicating 1:1 stoichiometry. Intermolecular hydrogen bonding involves the formation of SC, which does not undergo any chemical modification, followed by the complete conversion of the amorphous form which was identified by XRD. The in vitro cytotoxicity showed that IC50 was achieved in the SW480 (72 µM.mL−1) and Caco-2 (40 µM.mL−1) cells while that of pure CMN ranged from 146 to 116 µM/mL−1. Apoptosis studies showed that cell death is primarily due to apoptosis, with a low rate of necrosis. In vivo toxicity, confirmed by the zebrafish model, exhibited the safety of the HME-SC. In conclusion, the HME-SC potentially enhances the solubility and cytotoxicity to the treatment of colorectal cancer (CRC).
Collapse
|
43
|
Zhang H, Qu X, Han L, Di X. Mst2 Overexpression Inhibits Thyroid Carcinoma Growth and Metastasis by Disrupting Mitochondrial Fitness and Endoplasmic Reticulum Homeostasis. JOURNAL OF ONCOLOGY 2021; 2021:1262291. [PMID: 34557228 PMCID: PMC8455210 DOI: 10.1155/2021/1262291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 08/31/2021] [Indexed: 12/24/2022]
Abstract
Although the incidence of thyroid carcinoma has increased over the past several decades, it has an excellent prognosis and overall 5-year survival, with a stable mortality rate, except in cases with advanced stages or rare malignant tumor types. Biomarkers have emerged as effective targets of molecular therapy against thyroid carcinoma due to their rapid and convenient detection; however, there has been little clinical application. Macrophage stimulating 2 (Mst2) is a proapoptotic protein with implications in carcinogenesis and metastasis. We found that Mst2 overexpression-induced endoplasmic reticulum (ER) stress in MDA-T32 thyroid carcinoma cells, accompanied by elevated caspase-12 activity, increased apoptotic rate, and reduced cell viability. In addition, Mst2 overexpression contributed to mitochondrial damage, as evidenced by increased mitochondrial oxidative stress and activated the mitochondrial apoptotic pathway. Inhibition of the JNK pathway abolished these effects. These results show Mst2 to be a novel tumor suppressor that induces mitochondrial dysfunction and ER stress via the JNK pathway. Thus, Mst2 could potentially serve as a biomarker for developing targeted therapy against thyroid carcinoma.
Collapse
Affiliation(s)
- Haichao Zhang
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xin Qu
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Lu Han
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| | - Xu Di
- Department of Thyroid and Breast Surgery, Tianjin Fourth Central Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, China
| |
Collapse
|
44
|
Zhang L, Xu S, Cheng X, Wu J, Wang X, Wu L, Yu H, Bao J. Curcumin enhances the membrane trafficking of the sodium iodide symporter and augments radioiodine uptake in dedifferentiated thyroid cancer cells via suppression of the PI3K-AKT signaling pathway. Food Funct 2021; 12:8260-8273. [PMID: 34323243 DOI: 10.1039/d1fo01073e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Radioactive iodine (RAI) is commonly used to treat differentiated thyroid cancer (DTC). A major challenge is the dedifferentiation of DTC with the loss of radioiodine uptake. Patients with distant metastases have persistent or recurrent disease and develop resistance to RAI therapy due to tumor dedifferentiation. Hence, tumor redifferentiation to restore sensitivity to RAI therapy is considered a promising strategy to overcome RAI resistance. In the present study, curcumin, a natural polyphenolic compound, was found to re-induce cell differentiation and increase the expression of thyroid-specific transcription factors, TTF-1, TTF-2 and transcriptional factor paired box 8 (PAX8), and iodide-metabolizing proteins, including thyroid stimulating hormone receptor (TSHR), thyroid peroxidase (TPO) and sodium iodide symporter (NIS) in dedifferentiated thyroid cancer cell lines, BCPAP and KTC-1. Importantly, curcumin enhanced NIS glycosylation and its membrane trafficking, resulting in a significant improvement of radioiodine uptake in vitro. Additionally, AKT knockdown phenocopied the restoration of thyroid-specific gene expression; however, ectopic expressed AKT inhibited curcumin-induced up-regulation of NIS protein, demonstrating that curcumin might improve radioiodine sensitivity via the inhibition of the PI3K-AKT-mTOR signaling pathway. Our study demonstrates that curcumin could represent a promising adjunctive therapy for restoring iodide avidity and improve radioiodine therapeutic efficacy in patients with RAI-refractory thyroid carcinoma.
Collapse
Affiliation(s)
- Li Zhang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China. and Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China and School of Life Science and Technology, Southeast University, Nanjing 210096, China
| | - Shichen Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Xian Cheng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jing Wu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Xiaowen Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Liying Wu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Jiandong Bao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| |
Collapse
|
45
|
Bulotta S, Capriglione F, Celano M, Pecce V, Russo D, Maggisano V. Phytochemicals in thyroid cancer: analysis of the preclinical studies. Endocrine 2021; 73:8-15. [PMID: 33587255 DOI: 10.1007/s12020-021-02651-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/23/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE In the search for novel effective compounds to use in thyroid cancer (TC) unresponsive to current treatment, attention has recently focused on plant-derived compounds with anticancer activity. In this review, we discuss the preclinical studies demonstrating phytochemical activity against thyroid cancer cells. RESULTS/CONCLUSIONS In particular, we describe their antiproliferative properties or ability to re-induce iodine retention, thus supporting their potential use as single agents or adjuvants in radioiodine-resistant thyroid cancer treatment.
Collapse
Affiliation(s)
- Stefania Bulotta
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Viale Europa, Germaneto, 88100, Catanzaro, Italy
| | - Francesca Capriglione
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Viale Europa, Germaneto, 88100, Catanzaro, Italy
| | - Marilena Celano
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Viale Europa, Germaneto, 88100, Catanzaro, Italy
| | - Valeria Pecce
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico 155, 00161, Rome, Italy
| | - Diego Russo
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Viale Europa, Germaneto, 88100, Catanzaro, Italy.
| | - Valentina Maggisano
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Viale Europa, Germaneto, 88100, Catanzaro, Italy
| |
Collapse
|
46
|
Liang Y, Kong D, Zhang Y, Li S, Li Y, Dong L, Zhang N, Ma J. Curcumin inhibits the viability, migration and invasion of papillary thyroid cancer cells by regulating the miR-301a-3p/STAT3 axis. Exp Ther Med 2021; 22:875. [PMID: 34194553 PMCID: PMC8237388 DOI: 10.3892/etm.2021.10307] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Thyroid cancer is one of the most common malignant tumors, and the mortality rate associated with thyroid cancer has been increasing annually. Curcumin has been reported to exert an antitumor effect on papillary thyroid cancer (PTC), and the identification of additional mechanisms underlying the anticancer effect of curcumin on PTC requires further investigation. The present study aimed to explore the effects of curcumin on the viability, migration and invasion of PTC cells. TPC-1 cells were incubated with different concentrations of curcumin, and then, cell viability, migration and invasion, and wound healing were examined by CCK-8, Transwell and wound healing assays, respectively. Subsequently, microRNA (miR)-301a-3p mimics, miR-301a-3p inhibitors and signal transducer and activator of transcription (STAT)3 overexpression vector were transfected into TPC-1 cells, and cell viability, migration, and invasion were reassessed in these transfected cells. Matrix metallopeptidase (MMP)-2, MMP-9, epithelial-mesenchymal transition (EMT)-related markers, and Janus kinase (JAK)/STAT signaling pathway components were assessed by western blot analysis. Curcumin significantly inhibited cell viability, migration and invasion and downregulated MMP-2, MMP-9 and EMT marker expression. Additionally, curcumin decreased STAT3 expression by upregulating miR-301a-3p expression, and the inhibition of miR-301a-3p and the overexpression of STAT3 reversed the effects of curcumin on cell viability, migration and invasion, and MMP-2, MMP-9 and EMT marker expression in TPC-1 cells. Furthermore, curcumin suppressed the JAK/STAT signaling pathway through the miR-301a-3p/STAT3 axis. The data of the present study indicated that curcumin could inhibit the viability, migration and invasion of TPC-1 cells by regulating the miR-301a-3p/STAT3 axis. These findings may provide a possible strategy for the clinical treatment of PTC.
Collapse
Affiliation(s)
- Ying Liang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Deyu Kong
- Department of Internal Medicine-Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yi Zhang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Siqi Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Yan Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Liying Dong
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ningxin Zhang
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Junfeng Ma
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
47
|
Möller K, Macaulay B, Bein T. Curcumin Encapsulated in Crosslinked Cyclodextrin Nanoparticles Enables Immediate Inhibition of Cell Growth and Efficient Killing of Cancer Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:489. [PMID: 33672006 PMCID: PMC7919290 DOI: 10.3390/nano11020489] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022]
Abstract
The efficiency of anti-cancer drugs is commonly determined by endpoint assays after extended incubation times, often after days. Here we demonstrate that curcumin encapsulated in crosslinked cyclodextrin nanoparticles (CD-NP) acts extremely rapidly on cell metabolism resulting in an immediate and complete inhibition of cell growth and in efficient cancer-cell killing only few hours after incubation. This early onset of anti-cancer action was discovered by live-cell high-throughput fluorescence microscopy using an environmental stage. To date, only very few examples of covalently crosslinked nanoscale CD-based (CD-NP) drug carriers exist. Crosslinking cyclodextrins enables the adsorption of unusually high payloads of hydrophobic curcumin (762 µg CC/mg CD-NP) reflecting a molar ratio of 2.3:1 curcumin to cyclodextrin. We have investigated the effect of CD-NP encapsulated curcumin (CD-CC-NP) in comparison to free, DMSO-derived curcumin nanoparticles (CC-NP) on 4 different cell lines. Very short incubations times as low as 1 h were applied and cell responses after medium change were subsequently followed over two days. We show that cell proliferation is inhibited nearly immediately in all cell lines and that a cell- and concentration dependent cancer-cell killing occurs. Anti-cancer effects were similar with free and encapsulated curcumin, however, encapsulation in CD-NP drastically extends the long-term photostability and anti-cancer activity of curcumin. Curcumin-sensitivity is highest in HeLa cells reaching up to 90% cell death under these conditions. Sensitivity decreased from HeLa to T24 to MDA MB-231 cells. Strikingly, the immortalized non-cancerous cell line MCF-10A was robust against curcumin concentrations that were highly toxic to the other cell lines. Our results underline the potential of curcumin as gentle and yet effective natural anti-cancer agent when delivered solvent-free in stabilizing and biocompatible drug carriers such as CD-NP that enable efficient cellular delivery.
Collapse
Affiliation(s)
- Karin Möller
- Department of Chemistry and Center for NanoScience, University of Munich (LMU), Butenandtstrasse 5–13, 81377 Munich, Germany;
| | | | - Thomas Bein
- Department of Chemistry and Center for NanoScience, University of Munich (LMU), Butenandtstrasse 5–13, 81377 Munich, Germany;
| |
Collapse
|
48
|
Yang Q, Leong SA, Chan KP, Yuan XL, Ng TK. Complex effect of continuous curcumin exposure on human bone marrow-derived mesenchymal stem cell regenerative properties through matrix metalloproteinase regulation. Basic Clin Pharmacol Toxicol 2021; 128:141-153. [PMID: 32777138 DOI: 10.1111/bcpt.13477] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/13/2020] [Accepted: 08/04/2020] [Indexed: 02/05/2023]
Abstract
Curcumin has been reported to be beneficial for cancers, cardiovascular and neurodegenerative diseases, based on its anti-oxidative, anti-inflammation, anti-tumorigenic and neuroprotective properties. With its high-dose application, curcumin toxicity to systemic tissues is a reasonable concern. Here, we report the responses of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) to continuous curcumin exposure. hBM-MSCs were treated with 0.01-100 μmol/L curcumin continuously in vitro for 7 days. 25 μmol/L curcumin or above significantly attenuated hBM-MSC maintenance, whereas 10 μmol/L curcumin reduced hBM-MSC proliferation and hindered their migration with increasing cell apoptosis. Besides, 5 μmol/L curcumin treatment inhibited hBM-MSC adipogenic differentiation, but enhanced osteogenic differentiation, which depended on matrix metalloproteinase (MMP)-13 expression and activity. Furthermore, curcumin treatment reduced MMP1 expression but up-regulated the immunomodulatory gene IDO1 expression. In summary, this study revealed the complex effects of continuous curcumin exposure on hBM-MSC maintenance and regenerative properties through MMP regulation. Given the complex effects of curcumin, its use for biomedical purposes should be carefully considered in treatment length and dosage.
Collapse
Affiliation(s)
- Qichen Yang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Samantha Antonio Leong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Kwok Ping Chan
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Xiang-Ling Yuan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Tsz Kin Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
49
|
Olivas-Aguirre M, Torres-López L, Pottosin I, Dobrovinskaya O. Phenolic Compounds Cannabidiol, Curcumin and Quercetin Cause Mitochondrial Dysfunction and Suppress Acute Lymphoblastic Leukemia Cells. Int J Mol Sci 2020; 22:E204. [PMID: 33379175 PMCID: PMC7795267 DOI: 10.3390/ijms22010204] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Anticancer activity of different phenols is documented, but underlying mechanisms remain elusive. Recently, we have shown that cannabidiol kills the cells of acute lymphoblastic leukemia (ALL) by a direct interaction with mitochondria, with their consequent dysfunction. In the present study, cytotoxic effects of several phenolic compounds against human the T-ALL cell line Jurkat were tested by means of resazurin-based metabolic assay. To unravel underlying mechanisms, mitochondrial membrane potential (∆Ψm) and [Ca2+]m measurements were undertaken, and reactive oxygen species generation and cell death were evaluated by flow cytometry. Three out of eight tested phenolics, cannabidiol, curcumin and quercetin, which displayed a significant cytotoxic effect, also dissipated the ∆Ψm and induced a significant [Ca2+]m increase, whereas inefficient phenols did not. Dissipation of the ∆Ψm by cannabidiol was prevented by cyclosporine A and reverted by Ru360, inhibitors of the permeation transition pore and mitochondrial Ca2+ uniporter, respectively. Ru360 prevented the phenol-induced [Ca2+]m rise, but neither cyclosporine A nor Ru360 affected the curcumin- and quercetin-induced ∆Ψm depolarization. Ru360 impeded the curcumin- and cannabidiol-induced cell death. Thus, all three phenols exert their antileukemic activity via mitochondrial Ca2+ overload, whereas curcumin and quercetin suppress the metabolism of leukemic cells by direct mitochondrial uncoupling.
Collapse
Affiliation(s)
| | | | - Igor Pottosin
- Laboratory of Immunobiology and Ionic Transport Regulation, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Villa de San Sebastián, 28045 Colima, Mexico; (M.O.-A.); (L.T.-L.)
| | - Oxana Dobrovinskaya
- Laboratory of Immunobiology and Ionic Transport Regulation, Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de Julio 965, Villa de San Sebastián, 28045 Colima, Mexico; (M.O.-A.); (L.T.-L.)
| |
Collapse
|
50
|
Zhu G, Shen Q, Jiang H, Ji O, Zhu L, Zhang L. Curcumin inhibited the growth and invasion of human monocytic leukaemia SHI-1 cells in vivo by altering MAPK and MMP signalling. PHARMACEUTICAL BIOLOGY 2020; 58:25-34. [PMID: 31854220 PMCID: PMC6968541 DOI: 10.1080/13880209.2019.1701042] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/16/2019] [Accepted: 11/30/2019] [Indexed: 05/20/2023]
Abstract
Context: Curcumin, a polyphenolic compound extracted from the rhizome of the tropical plant Curcuma longa L. (Zingiberaceae), has been considered as a cancer chemopreventive drug by American National Cancer Institute.Objective: To examine the effect of curcumin on acute monocytic leukaemia SHI-1 cells in vivo.Materials and methods: The SHI-1 cells (1 × 106 cells in 0.1 mL PBS) were injected subcutaneously into the right flanks of the female SCID mice. Curcumin dissolved in olive oil (15 and 30 mg/kg) was administered (i.p.) to mice once a day for 15 days while the control group received olive oil injection. Tumour proliferation and apoptosis were examined by PCNA, TUNEL and cleaved caspase-3 staining. The expression of MAPK, NF-κB, MMP9, MMP2 and vimentin were confirmed by RT-PCR, immunohistochemistry or western blotting.Results: Administration of curcumin significantly inhibited tumour growth, as the tumour weight decreased from 0.67 g (control) to 0.47 g (15 mg/kg) and 0.35 g (30 mg/kg). Curcumin inhibited the expression of PCNA and increased the degree of TUNEL and cleaved caspase-3 staining in tumour tissue. The results of western blotting showed that curcumin treatment inhibited NF-κB and ERK signalling while activating p38 and JNK. Moreover, curcumin attenuated the mRNA transcription and protein expression of MMP2 and MMP9. Curcumin also suppressed the level of vimentin.Discussion and conclusions: Our study demonstrates that curcumin can inhibit the growth and invasion of human monocytic leukaemia in vivo, suggesting the possible use of curcumin for anti-metastasis in leukaemia and the value of determining its unique target.
Collapse
Affiliation(s)
- Guohua Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
- CONTACT Guohua Zhu First Clinical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qun Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, First Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Jiang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ou Ji
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Linyang Zhang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|