1
|
Durmaz A, Mizan Kahyaoğlu İ, Can Aytar E, Danışman Kalındemirtaş F, Sert E, Erol‐Bozkurt A, Karakuş S. In Vitro, Anti-Colon Cancer Activity of Green Dumbbell-Shaped Rhododendron luteum-Based Carbon Dots. ChemistryOpen 2025; 14:e202400303. [PMID: 39888200 PMCID: PMC12075102 DOI: 10.1002/open.202400303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/08/2025] [Indexed: 02/01/2025] Open
Abstract
Colorectal cancer is the second most common cause of cancer-related deaths worldwide and the third most common cancer overall. In this study, we investigate the anti-colon cancer potential of phytochemically, and thermally synthesised novel green carbon dots based on Rhododendron luteum (RL-CDs). A new synthesis method was used to produce carbon dots obtained from the Rhododendron luteum (RL) plant in an environmentally friendly manner. The green RL were characterized using Fourier-transform infrared spectroscopy (FTIR), UV-Vis spectroscopy, transmission electron microscopy (TEM), and artificial intelligence (AI)-based TEM analysis. The FTIR spectrum showed peaks corresponding to the hydroxyl (-OH) vibration of polyphenols at 3500 cm-1, the C=O vibration of cellulose derivatives at 1728 cm-1, and the C-O stretching of primary alcohol at 1041 cm-1. Two UV absorption peaks at roughly 253 nm (UV-C range), and 320 nm (UV-B range) were observed. The size of the green RL was measured to be less than 50 nm, and its morphology was characterized as dumbbell-shaped through TEM analysis. In-vitro studies were performed with HCT116 colon cancer, MCF-7 breast cancer, and normal HUVEC cells. The results demonstrated that the RL-CDs exhibited selective cytotoxic activity against HCT116 colon cancer cells. The results show that the RL extract stimulates cancer cell death by decreasing the CD44/24 ratio, and increasing apoptotic activity. These observations suggest that green RL-CDs could be an effective anticancer agent in colon cancer therapy, investigating their potential in this direction could be a promising way for future research.
Collapse
Affiliation(s)
- Alper Durmaz
- Ali Nihat Gökyiğit Botanical Garden Application and Research CenterArtvin Çoruh University08000ArtvinTürkiye
| | | | - Erdi Can Aytar
- Department of HorticultureFaculty of AgricultureUsak University64200UsakTürkiye
| | | | - Esra Sert
- Department of HematologyIstanbul Faculty of MedicineIstanbul University34093IstanbulTürkiye
| | - Ayşe Erol‐Bozkurt
- Department of Medical BiologyIstanbul Faculty of MedicineIstanbul University34093IstanbulTürkiye
| | - Selcan Karakuş
- Department of ChemistryFaculty of EngineeringIstanbul University-Cerrahpaşa34320IstanbulTürkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Esenler34220IstanbulTürkiye
| |
Collapse
|
2
|
Ozdemir N, Tan G, Tevlek A, Arslan G, Zengin G, Sargin I. Dead Cell Discrimination with Red Emissive Carbon Quantum Dots from the Medicinal and Edible Herb Echinophora tenuifolia. J Fluoresc 2025:10.1007/s10895-025-04286-y. [PMID: 40186814 DOI: 10.1007/s10895-025-04286-y] [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: 02/04/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
Accurately determining the viability of cells is crucial for in vitro cell research. Fluorescence-based live/dead cell staining is a highly desirable method to assess cell viability and survival in in vitro studies. We describe a green synthesis method to create red-emissive CQDs from the medicinal and edible herb Echinophora tenuifolia using microwave irradiation. We observed that the biocompatibility and photostability of the CQDs are superior. The antioxidant capacity of the CQDs and the plant extract were also investigated using different chemical methods (DPPH, ABTS, CUPRAC, FRAP, PBD, and MCA). The antioxidant capacity of the CQDs was similar to that of the extract of E. tenuifolia. Cytotoxicity studies indicate that while the CQDs are not toxic to L929, they exhibit significant toxicity towards HepG2 cells. The CQDs exhibited a strong negative zeta potential (-44.0 mV), which contributed to their selective interaction with dead cells while being repelled by viable cells with intact membrane potentials. The optimal concentration for effective, non-toxic imaging was determined to be 25 µg/mL, as lower concentrations did not produce detectable fluorescence. Differential staining experiments confirmed that CQDs selectively stained dead cells, with red fluorescence observed under the Texas Red filter. Moreover, CQDs exhibited favorable fluorescence intensity and stability, which may offer advantages for long-term and reliable bioimaging applications. In vitro studies on HepG2 and L929 cell lines revealed that the red-emissive CQDs from E. tenuifolia can be potentially used in bioimaging.
Collapse
Affiliation(s)
- Naciye Ozdemir
- Department of Biochemistry, Faculty of Science, Selçuk University, Konya, 42075, Turkey
| | - Gamze Tan
- Department of Biology, Faculty of Science and Letters, Aksaray University, Aksaray, 68100, Turkey
| | - Atakan Tevlek
- Department of Medical Biology, Faculty of Medicine, Atilim University, Ankara, 06830, Turkey
| | - Gulsin Arslan
- Department of Biochemistry, Faculty of Science, Selçuk University, Konya, 42075, Turkey
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selçuk University, Konya, 42075, Turkey
| | - Idris Sargin
- Department of Biochemistry, Faculty of Science, Selçuk University, Konya, 42075, Turkey.
| |
Collapse
|
3
|
Wang Y, Yang Z, Li Z, Huang L, Hou S, Wang J, Yu Y, Yin J, Wu J. Proteomics-driven discovery of LCAT as a novel biomarker for liver metastasis in colorectal cancer. BMC Cancer 2025; 25:480. [PMID: 40089671 PMCID: PMC11909834 DOI: 10.1186/s12885-025-13882-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND This study aimed to identify molecular markers that influence liver metastasis in colorectal cancer (CRC) and assess their clinical relevance. METHODS Proteomic analysis compared differential protein expression between CRC patients with liver metastasis (CRLM) and those without (CRNLM). Bioinformatics and survival analyses identified key proteins and validated them using the TCGA database for expression and clinical significance. Clinical and pathological data, along with tissue samples from our center, were used to create tissue microarrays for immunohistochemistry. Logistic regression assessed odds ratios (OR) for molecular markers linked to liver metastasis post-CRC surgery. Stable LCAT knockdown and overexpression CRC cell lines were constructed, and Transwell assays assessed the impact LCAT on cell migration. Nile red staining of these cells validated the effect LCAT on lipid metabolism in CRC cells. RESULTS Proteomic analysis identified 383 differentially expressed proteins between the CRLM and CRNLM groups (212 upregulated, 171 downregulated). Enrichment analysis linked these proteins to steroid and alcohol metabolism, inflammation, lipoproteins, and HDL particles, with key pathways in cholesterol and retinol metabolism. Lecithin cholesterol acyltransferase (LCAT), an important enzyme in this process, showed higher expression in CRC tissues, with increased LCAT linked to poorer 5-year OS, DSS, and PFI. LCAT expression also increased with tumor stage. Among 119 patients with CRC, preoperative complications, tumor staging, and LCAT scores differed significantly between patients with and without liver metastasis within 3 years post-surgery. LCAT and postoperative CEA levels were independent risk factors for liver metastasis (LCAT OR, 10.221; P = 0.002; CEA OR, 1.296; P = 0.014). Western blotting confirmed significantly higher LCAT expression in CRC tissues with liver metastasis. Transwell assays showed that LCAT overexpression enhanced migratory ability, while knockdown inhibited it. Nile red staining revealed increased lipid droplet accumulation in LCAT-overexpressing CRC cells, which was reduced by LCAT knockdown. CONCLUSION LCAT, which is involved in lipid metabolism, is an independent risk factor for liver metastasis following CRC surgery, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yuyao Wang
- Dalian Medical University, Dalian, China
- Department of Gastrointestinal Surgery, Chengdu Sixth People's Hospital, Chengdu, China
| | - Zhengbo Yang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China
| | - Ziqun Li
- Dalian Medical University, Dalian, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China
| | - Linglong Huang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China
| | - Shuangshuang Hou
- Department of Surgery, Fuyang Normal University Second Affiliated Hospital, Fuyang, China
| | - Jiaqi Wang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China
| | - Yang Yu
- Dalian Medical University, Dalian, China
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China
| | - Jiajun Yin
- Dalian Medical University, Dalian, China.
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China.
| | - Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Zhongshan District, Dalian, 116001, Liaoning Province, China.
| |
Collapse
|
4
|
Feng S, Li S, Wu Z, Li Y, Wu T, Zhou Z, Liu X, Chen J, Fu S, Wang Z, Zhong Z, Zhong Y. Saffron improves the efficacy of immunotherapy for colorectal cancer through the IL-17 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118854. [PMID: 39326815 DOI: 10.1016/j.jep.2024.118854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/08/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saffron is one of the traditional medicinal herbs, which contains various active ingredients, such as safranal, crocin, saffron acid, etc. It has anti-inflammatory, antioxidant, and anti-cancer properties, and is widely used in clinical practice. The anti-cancer efficacy of saffron has been previously confirmed, but its anti-cancer mechanism in colorectal cancer remains unclear. OBJECTIVE We investigated the effect of active compounds of saffron on the efficacy of immunotherapy for colorectal cancer. METHODS TCMSP and liquid chromatography-mass spectrometry analysis (LC-MS), GeneCards, and DisGeNET databases were used to identify the active compounds of saffron, drug targets and the disease targets of colorectal cancer. They were then subjected to Gene Ontology Enrichment (GO) and Signalling Pathway Enrichment (KEGG) analyses. The core targets and corresponding compounds were selected for molecular docking. The effect of active components of saffron on the proliferation of CT26 and HCT116 cells was investigated using the cell counting kit-8 (CCK-8). In vitro experiments were conducted by subcutaneous injection of CT26 cells to establish a colon cancer model. Enzyme-linked immunosorbent assay (ELISA), western blotting (WB), real-time polymerase chain reaction (RT-PCR), immunohistochemistry (IHC), and flow cytometry (FCM) were employed to validate the effects of saffron on colorectal cancer immunotherapy. RESULTS 1. LC-MS analysis revealed that the main active component of saffron extract was crocin. The active chemicals of saffron intersected with 170 colorectal cancer targets, with 17 predicting targets for saffron treatment. GO and KEGG enrichment analyses revealed that the active components of saffron can prevent colorectal cancer development by enhancing Th17 cell differentiation and the IL-17 signaling pathway. 2. In vitro studies revealed that saffron alcohol extract, crocin, and safranal can suppress the proliferation of CT26 and HCT116 cells. 3. In vivo studies showed that crocin and safranal can increase the body mass and decrease the tumor mass of loaded mice, decrease the serum level of IL-17, and lower the mRNA expression level of IL-17, IL-6, TNF-α, TGF-β, and PD-L1 and IL-17, PD-L1 protein in tumors. This inhibitory effect was strengthened after combined immunotherapy. In addition, saffron modulated CD4+ and CD8+ T cells and the CD4+/CD8+T ratio in mouse spleens. CONCLUSION The active components of saffron can reduce the expression of inflammatory factors and ameliorate the immunological microenvironment of tumors via the IL-17 signaling pathway, thereby improving the efficacy of immunotherapy for colorectal cancer. This study provides pharmacological support for the application of saffron in enhancing the efficacy of immunotherapy for colorectal cancer.
Collapse
Affiliation(s)
- Siqi Feng
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Shiying Li
- Seoul National University, Seoul, Korea.
| | - Zhonghua Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yun Li
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Tingting Wu
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Zhangjie Zhou
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Xinhua Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jian Chen
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Shujuan Fu
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| | - Zhiying Wang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | | | - Yi Zhong
- Shanghai TCM-integrated Hospital, Shanghai University of TCM, Shanghai, China.
| |
Collapse
|
5
|
Feng Q, Sun Y, Yang Z, Wang Z, Chen Z, Liu F, Liu L. Copper in the colorectal cancer microenvironment: pioneering a new era of cuproptosis-based therapy. Front Oncol 2025; 14:1522919. [PMID: 39850821 PMCID: PMC11754209 DOI: 10.3389/fonc.2024.1522919] [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: 11/05/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Copper, an essential trace element and biochemical cofactor in humans plays a critical role in maintaining health. Recent studies have identified a significant association between copper levels and the progression and metastasis of cancer. Copper is primarily absorbed in the intestinal tract, often leading to an imbalance of copper ions in the body. Colorectal cancer (CRC), the most common cancer originating in the intestines, thrives in an environment with elevated copper concentrations. Current research is focused on uncovering the relationship between copper and CRC which has introduced new concepts such as cuproplasia and cuproptosis, significantly deepening our understanding of copper's influence on cell proliferation and death. Cuproplasia is a kind of cell proliferation mediated by the co-regulatory activities of enzymes and non-enzymatic factors, while cuproptosis refers to cell death induced by excessive copper, which results in abnormal oligomerization of lipacylated proteins and the reduction of iron-sulfur cluster proteins. Exploring cuproplasia and cuproptosis opens new avenues for treating CRC. This review aims to summarize the critical role of copper in promoting colorectal cancer, the dual effects of copper in the tumor microenvironment (TME), and strategies for leveraging this unique microenvironment to induce cuproptosis in colorectal cancer. Understanding the relationship between copper and CRC holds promise for establishing a theoretical foundation for innovative therapeutic strategies in CRC.
Collapse
Affiliation(s)
- Qixuan Feng
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhe Yang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiyu Wang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhangyi Chen
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fang Liu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Jing HH, Shati AA, Alfaifi MY, Elbehairi SEI, Sasidharan S. The future of plant based green carbon dots as cancer Nanomedicine: From current progress to future Perspectives and beyond. J Adv Res 2025; 67:133-159. [PMID: 38320729 PMCID: PMC11725112 DOI: 10.1016/j.jare.2024.01.034] [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: 09/20/2023] [Revised: 12/18/2023] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The emergence of carbon dots (CDs) as anticancer agents had sparked a transformation in cancer research and treatment strategies. These fluorescent CDs, initially introduced in the early 2000 s, possess exceptional biocompatibility, tunable fluorescence, and surface modification capabilities, positioning them as promising tools in biomedical applications. AIM OF REVIEW The review encapsulates the transformative trajectory of green CDs as future anticancer nanomedicine, poised to redefine the strategies employed in the ongoing fight against cancer. KEY SCIENTIFIC CONCEPTS OF REVIEW The versatility of CDs was rooted in their various synthesis approaches and sustainable strategies, enabling their adaptability for diverse therapeutic uses. In vitro studies had showcased CDs' selective cytotoxicity against cancer cells while sparing healthy counterparts, forming the basis for targeted therapeutic potential. This selectivity had been attributed to the reactive oxygen species (ROS) generation, which opened avenues for targeted interventions. The role of CDs in combination therapies, synergizing with chemotherapy, radiotherapy, and targeted approaches was then investigated to heighten their anticancer efficacy. Notably, in vivo studies highlight CDs' remarkable biocompatibility and minimal side effects, endorsing their translational promise. Integration with conventional cancer treatments such as chemotherapy, radiotherapy, and immunotherapy amplified the versatility and effectiveness of CDs. The exploration of CDs' applications in photo-induced treatments further solidified their significance, positioning them as photosensitizers (PS) in photodynamic therapy (PDT) and photothermal agents (PA) in photothermal therapy (PTT). In PDT, CDs triggered the generation of ROS upon light exposure, facilitating cancer cell elimination, while in PTT, they induced localized hyperthermia within cancer cells, enhancing therapeutic outcomes. In vitro and in vivo investigations validated CDs' efficacy in PDT and PTT, affirming their potential for integration into combination therapies. Looking ahead, the future of CDs in anticancer treatment encompasses bioavailability, biocompatibility, synergistic treatments, tumor targeting, artificial intelligence (AI) and robotics integration, personalized medicine, and clinical translation. This transformative odyssey of CDs as future anticancer agents is poised to redefine the paradigm of cancer treatment strategies.
Collapse
Affiliation(s)
- Hong Hui Jing
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Pulau Pinang 11800, Malaysia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia; Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), 51 Wezaret El-Zeraa St., Agouza, Giza, Egypt
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia (USM), Pulau Pinang 11800, Malaysia.
| |
Collapse
|
7
|
Hallaj T, Soraya H, Abbasi A, Tasoujlu M, Pirmoradi S, Ansari L. Chitosan gel loaded with carbon dots and mesoporous hydroxyapatite nanoparticles as a topical formulation for skin regeneration: An animal study. Int J Pharm 2024; 666:124828. [PMID: 39414185 DOI: 10.1016/j.ijpharm.2024.124828] [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: 06/01/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND Carbon dots (CDs) are the research hotspots in antimicrobial applications. Also, due to the importance of calcium ions in skin regeneration, mesoporous hydroxyapatite nanoparticles (mHAP NPs) were prepared. This study aimed to evaluate the wound healing efficiency of both CD and mHAP NPs simultaneously. METHODS We doped Fluorine (F) and Copper (Cu) in CDs to prepare Cu*F-doped CDs (Cu.CDs). For the rapid release of calcium ions, a novel double-surfactant system (CTAB/F-127) was used for the synthesis of mHAP NPs. Then, we loaded both Cu.CDs and mHAP NPs simultaneously into the chitosan (CTS) gel. RESULTS The released calcium for mHAP NPs prepared using CTAB/F-127 was proved about 30 % higher than that prepared using only CTAB, after one day. The BET-specific surface area for mHAP NPs prepared using CTAB/F-127 was 23.17 % more than that prepared using only CTAB. This high porosity improves wound exudate absorption. Rheological analysis showed that the addition of both mHAP NPs and Cu.CD decreases the viscosity of CTS gel and thereby makes it suitable for topical applications. CTS gel containing mHAP + Cu.CDs indicated exceptional antibacterial properties even without light and/or H2O2 with an inhibition zone diameter of 35 and 17 mm for E. coli and S. aureus species, respectively. CTS gel containing mHAP + Cu.CDs also showed significantly higher wound closure efficacy, greater inflammation inhibition, and better re-epithelialization than the other groups in rats with full-thickness skin wounds. CONCLUSION The CTS gel containing both Cu.CD and mHAP NPs can be proposed as a new therapeutic gel for wound healing.
Collapse
Affiliation(s)
- Tooba Hallaj
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hamid Soraya
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Ata Abbasi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Pathology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mina Tasoujlu
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Samaneh Pirmoradi
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Legha Ansari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
8
|
Ebrahimi Y, Rezaie J, Akbari A, Rasmi Y. Green hydrothermal synthesis of gallic acid carbon dots: characterization and cytotoxic effects on colorectal cancer cell line. Biomed Phys Eng Express 2024; 11:015017. [PMID: 39530648 DOI: 10.1088/2057-1976/ad9153] [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: 07/17/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024]
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide, necessitating the development of novel therapeutic approaches. Carbon dots (CDs) have emerged as promising nanoparticles for biomedical applications due to their unique properties. Gallic acid (GA), an anticancer agent, is effective against various tumor types. This study explores the potential of gallic acid-derived carbon dots (GA-CDs) as an innovative anticancer agent against HCT-116 CRC cells, focusing on apoptosis signaling pathways. GA-CDs were synthesized using a one-pot hydrothermal method. Characterization was conducted using transmission electron microscopy (TEM), Fourier transform infrared (FT-IR) spectroscopy, and ultraviolet-visible (UV-vis) absorption spectroscopy. The cytotoxicity of GA and GA-CDs on HCT-116 cells was evaluated using the MTT assay at various concentrations over 24 and 48 h. Cellular uptake was assessed via fluorescence microscopy, and apoptosis was analyzed using acridine orange/propidium iodide (AO/PI) staining. Total RNA extraction followed by complementary DNA (cDNA) synthesis via reverse transcription-PCR was performed, and real time-PCR (Q-PCR) was conducted to examine the expression of apoptosis-related genes includingCaspase-3,Bax, andBcl-2. Characterization confirmed the successful synthesis of spherical GA-CDs. GA-CDs exhibited dose- and time-dependent cytotoxicity, with IC50 values of 88.55 μg ml-1for GA-CDs and 192.2 μg ml-1for GA after 24 h. Fluorescence microscopy confirmed the efficient uptake of GA-CDs by HCT-116 cells. AO/PI staining showed a significant increase in apoptotic cell numbers after treatment with GA-CDs. Q-PCR analysis revealed overexpression ofCaspase-3 andBaxgenes in GA-CD-treated cells, though no significant changes were observed in the expression ofBcl-2 or theBax/Bcl-2 ratio. GA-CDs demonstrated potent anticancer properties by inducing apoptosis and reducing cell viability in HCT-116 cells. These findings suggest the potential of GA-CDs as a novel therapeutic agent for CRC treatment, warranting further investigation into their mechanism of action andin vivoefficacy.
Collapse
Affiliation(s)
- Yaser Ebrahimi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Akbari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
9
|
Akbari A, Nemati M, Lighvan ZM, Khanamiri FN, Rezaie J, Rasmi Y. Synthesis of metformin-derived fluorescent quantum dots: uptake, cytotoxicity, and inhibition in human breast cancer cells through autophagy pathway. J Biol Eng 2024; 18:38. [PMID: 38915025 PMCID: PMC11197241 DOI: 10.1186/s13036-024-00433-4] [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: 04/14/2024] [Accepted: 06/13/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Breast cancer remains a challenge for physicians. Metformin, an antidiabetic drug, show promising anticancer properties against cancers. An emerging quantum dot (QD) material improves therapeutic agents' anticancer and imaging properties. QD are nano-sized particles with extreme application in nanotechnology captured by cells and accumulated inside cells, suggesting bioimaging and effective anticancer outcomes. In this study, a simple one-pot hydrothermal method was used to synthesize fluorescent metformin-derived carbon dots (M-CDs) and then investigated the cytotoxic effects and imaging features on two human breast cancer cell lines including, MCF-7 and MDA-MB-231 cells. RESULTS Results showed that M-CDs profoundly decreased the viability of both cancer cells. IC50 values showed that M-CDs were more cytotoxic than metformin either 24-48 h post-treatment. Cancer cells uptake M-CDs successfully, which causes morphological changes in cells and increased levels of intracellular ROS. The number of Oil Red O-positive cells and the expression of caspase-3 protein were increased in M-CDs treated cells. Authophagic factors including, AMPK, mTOR, and P62 were down-regulated, while p-AMPK, Becline-1, LC3 I, and LC3 II were up-regulated in M-CDs treated cells. Finally, M-CDs caused a decrease in the wound healing rate of cells. CONCLUSIONS For the first, M-CDs were synthesized by simple one-pot hydrothermal treatment without further purification. M-CDs inhibited both breast cancer cells through modulating autophagy signalling.
Collapse
Affiliation(s)
- Ali Akbari
- Solid Tumor Research Center, Cellular and Molecular Research Medicine Institute, Urmia University of Medical Sciences, P.O. BoX: 1138, Shafa St, Ershad Blvd, Urmia, 57147, Iran
| | - Mohadeseh Nemati
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zohreh Mehri Lighvan
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, P.O. Box 14965-115, Tehran, Iran
| | - Fereshteh Nazari Khanamiri
- Solid Tumor Research Center, Cellular and Molecular Research Medicine Institute, Urmia University of Medical Sciences, P.O. BoX: 1138, Shafa St, Ershad Blvd, Urmia, 57147, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Research Medicine Institute, Urmia University of Medical Sciences, P.O. BoX: 1138, Shafa St, Ershad Blvd, Urmia, 57147, Iran.
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
10
|
Bokhari SS, Ali T, Naeem M, Hussain F, Nasir A. Recent advances in nanoformulation-based delivery for cancer immunotherapy. Nanomedicine (Lond) 2024; 19:1253-1269. [PMID: 38717427 PMCID: PMC11285355 DOI: 10.1080/17435889.2024.2343273] [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: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 07/25/2024] Open
Abstract
Cancer is one of the leading causes of mortality worldwide, and its treatment faces several challenges. Phytoconstituents derived from recently discovered medicinal plants through nanotechnology potentially target cancer cells via PI3K/Akt/mTOR pathways and exert their effects selectively through the generation of reactive oxygen species through β-catenin inhibition, DNA damage, and increasing caspase 3/9 and p53 expression. These nanocarriers act specifically against different cancer cell lines such as HT-29, MOLT-4 human leukemia cancer and MCF-7 cell lines SKOV-3, Caov-3, SW-626, HepG2, A-549, HeLa, and MCF-7. This review comprehensively elaborates on the cellular and molecular mechanisms, and therapeutic prospects of various plant-mediated nanoformulations to attain a revolutionary shift in cancer immunotherapy.
Collapse
Affiliation(s)
- Seyedeh Saimeh Bokhari
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Tayyab Ali
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Fatma Hussain
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Abdul Nasir
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
11
|
Manimaran K, Yuli Yanto DH, Kamaraj C, Selvaraj K, Pandiaraj S, M Elgorban A, Vignesh S, Kim H. Eco-friendly approaches of mycosynthesized copper oxide nanoparticles (CuONPs) using Pleurotus citrinopileatus mushroom extracts and their biological applications. ENVIRONMENTAL RESEARCH 2023:116319. [PMID: 37271436 DOI: 10.1016/j.envres.2023.116319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/26/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
This current study aims to develop a unique biomaterial that can fight against oxidative stress and microbial infections without causing any harm. As a result, an easy-to-make, environment-friendly, long-lasting, and non-toxic copper oxide nanoparticle (CuONP) was synthesized using an edible mushroom Pleurotus citrinopileatus extract. The UV-visa spectroscopy analyses reflected a sharp absorbance peak at 250 nm. The FTIR, XRD, SEM, HR-TEM, and EDX instrumental tools were used to characterize the myco-produced CuONPs. The face-centred cubic (FCC) CuONPs were found to have diffraction peaks at the planes of (110), (002), (111), (112), (020), (202), (113), (310), (220), and (004). The HR-TEM result showed the particles having a spherical structure and an average nanoparticles size of 20 nm. The antimicrobial activity results expressed the broad spectrum of antibacterial effect and the better growth inhibition zone was recorded in P. aeruginosa (8.3 ± 0.1), E. coli (7.4 ± 0.3), K. pneumoniae (7.2 ± 0.1), S. aureus (7.1 ± 0.3), S. pneumoniae (6.3 ± 0.2), and B. cereus (6.2 ± 0.3). The cytotoxicity efficacy of myco-synthesized CuONPs tested against a cancer cell line (HT-29) observed the best result in low doses of mushroom extract (45.62 μg/mL). Based on the outcome of the study suggests that the mycosynthesized CuONPs using Pleurotus mushroom extract might serve as an alternative agent for biomedical applications in the near future.
Collapse
Affiliation(s)
- Kumar Manimaran
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), JI. Raya Bogor KM. 46, Cibinong, 16911, Indonesia; Department of Product Development, Institute of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, 602105, India.
| | - Dede Heri Yuli Yanto
- Research Center for Applied Microbiology, National Research and Innovation Agency (BRIN), JI. Raya Bogor KM. 46, Cibinong, 16911, Indonesia.
| | - Chinnaperumal Kamaraj
- Interdisciplinary Institute of Indian System of Medicine (IIISM), Directorate of Research, SRM Institute of Science and Technology (SRM IST), Kattankulathur, 603203, Tamil Nadu, India
| | - Kumar Selvaraj
- Facultad de Ingeniería Química, Universidad Nacional del Litoral (UNL), Santiago del Estero 2829, Santa Fe, 3000, Argentina
| | - Saravanan Pandiaraj
- Department of Self-Development Skills, King Saud University, P.O. 2455, Riyadh, 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. 2455, Riyadh, 11451, Saudi Arabia
| | - Shanmugam Vignesh
- School of Materials Science and Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| | - Haekyoung Kim
- School of Materials Science and Engineering, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|