1
|
Song Y, Zhao QL, Ogawa R, Mizukami T, Li YM, Cui ZG, Saitoh JI, Noguchi K. Exploring the therapeutic potential of 4,4'-dimethoxychalcone: Inducing apoptosis in cancer cells via ER stress and autophagy disruption. Cell Signal 2025; 132:111854. [PMID: 40334804 DOI: 10.1016/j.cellsig.2025.111854] [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: 01/27/2025] [Revised: 04/10/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
In cancer therapeutics, natural flavonoid compounds are renowned for their diverse structures and broad biological activities, offering considerable opportunities for drug discovery. This study investigates the anticancer effects of the flavonoid 4,4'-dimethoxychalcone (DMC), focusing on its apoptotic mechanisms and therapeutic potential. Our findings reveal that DMC induces apoptosis by upregulating pro-apoptotic proteins (Bax, Bim, tBid) and downregulating anti-apoptotic proteins (Bcl-2, Mcl-1), with concurrent caspase-3 activation and PARP cleavage. This apoptotic effect is mitigated by Z-VAD-FMK, a pan-caspase inhibitor. DMC also induces mitochondrial membrane potential (MMP) loss and increases reactive oxygen species (ROS) production. Furthermore, DMC promotes endoplasmic reticulum (ER) stress, evidenced by the increased expression of p-PERK/PERK, p-IRE1/IRE1, GRP78, HSP70, ATF4, and CHOP proteins. ER stress inhibitors significantly reverse DMC-induced MMP loss, apoptosis, and upregulation of apoptosis-related proteins. Additionally, DMC activates the mitogen-activated protein kinase (MAPK) pathway, including Erk, JNK, and p38. DMC also promotes autophagosome accumulation, modulates autophagy marker proteins (LC3-II, ATG5, p62), and leads to lysosomal dysfunction-evidenced by downregulated LAMP-1 and Cathepsin D expression, lysosomal pH increase, yet unaffected LC3 and LAMP-1 co-localization. Modulating autophagy with inhibitors (3-methyladenine, 3-MA; chloroquine, CQ) or an inducer (rapamycin, Rapa) respectively enhances or reduces DMC-induced apoptosis. Treatment with 3-MA also led to a significant increase in the expression of ER stress markers CHOP and ATF4. Collectively, DMC-induced cell death is primarily due to ER stress activation and autophagic flux impairment via lysosomal dysfunction. These results suggest DMC's potential as an anticancer agent, warranting further clinical investigation.
Collapse
Affiliation(s)
- Yu Song
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Qing-Li Zhao
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Ryohei Ogawa
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Tatsuji Mizukami
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Yu-Mei Li
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Zheng-Guo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, Fukui 910-1193, Japan.
| | - Jun-Ichi Saitoh
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan.
| | - Kyo Noguchi
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
2
|
Doan CC, Le TL, Ho NQC, Bui DT, Nguyen TPT, Hoang NS. Cytotoxicity evaluation of Curcuma aromatica Salisb. rhizome extract via apoptosis and reactive oxygen species generation in human gastric cancer cells. 3 Biotech 2025; 15:153. [PMID: 40342537 PMCID: PMC12055678 DOI: 10.1007/s13205-025-04318-1] [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: 07/01/2024] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Curcuma aromatica Salisb. (C. aromatica) has been reported to have anti-proliferative activities against different types of cancer, including colon, lung, skin, and prostate cancer. In this study, we aimed to determine whether C. aromatica rhizome extract (CAE) exhibited cytotoxic and apoptotic effects against gastric cancer cells. CAE exerted cytotoxic effects on different cancer cells by suppressing cell growth in a concentration-dependent manner, while its effects on normal cells were minimal at equivalent doses. The extract showed induction of apoptosis in gastric cancer AGS cells via activation of extrinsic apoptosis. This effect was associated with the upregulation of proteins related to death receptors, including Fas, FasL, DR5, FADD, and caspase-8. Additionally, it activated intrinsic apoptosis by causing mitochondrial dysfunction. This was linked to a downregulation of Bid expression, an increment in the Bax/Bcl-2 ratio, and a disruption of mitochondrial membrane potential (ΔΨm). As a result, cytochrome c was released into the cytoplasm, leading to activation of caspase-9 and -3, followed by cleavage of PARP. Moreover, blocking the activity of caspase-8, -9, and -3 by specific caspase inhibitors protected AGS cells against apoptosis by CAE treatment. Also, CAE significantly promoted cell death in AGS cells via an augmentation in ROS generation, which was attenuated by a potent antioxidant, NAC. Alternatively, curdione, curcumenol, and germacrone were found in CAE as potential active constituents. Our results suggested that CAE triggered apoptosis via the activation of both exogenous and endogenous apoptosis pathways and the enhancement of intracellular ROS generation in gastric cancer AGS cells. Thus, CAE might be a novel potential candidate for the treatment of gastric cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-025-04318-1.
Collapse
Affiliation(s)
- Chinh Chung Doan
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Thanh Long Le
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Nguyen Quynh Chi Ho
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Dinh Thach Bui
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Thi Phuong Thao Nguyen
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| | - Nghia Son Hoang
- Department of Animal Biotechnology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
- Faculty of Biotechnology, Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Ha Noi City, Vietnam
| |
Collapse
|
3
|
Lan Y, Sun J, Xu J, Chen X. Anti-lung cancer activity of lotusine in non-small cell lung cancer HCC827 via reducing proliferation, oxidative stress, induction of apoptosis, and G0/G1 cell cycle arrest via suppressing EGFR-Akt-ERK signalling. In Vitro Cell Dev Biol Anim 2025:10.1007/s11626-025-01048-9. [PMID: 40392483 DOI: 10.1007/s11626-025-01048-9] [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: 02/13/2025] [Accepted: 04/10/2025] [Indexed: 05/22/2025]
Abstract
Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related deaths worldwide, with resistance to targeted therapies and the need for novel therapeutic agents driving ongoing research. In this study, we investigated the anti-lung cancer activity of lotusine, a natural alkaloid, in the A549 (non-EGFR mutant), and EGFR-mutant HCC827 NSCLC cell line (deletion in exon 19). Our results demonstrated that lotusine significantly inhibited cell proliferation in a concentration- and time-dependent manner of HCC827 cells in comparison to A549 cells. Mechanistic analysis revealed that lotusine induced apoptosis in HCC827 cells, as evidenced by increased expression of pro-apoptotic markers (Bax and cleaved caspase-3) and decreased levels of anti-apoptotic proteins (Bcl-2). Cell cycle analysis indicated that lotusine caused G0/G1 phase arrest. Importantly, lotusine exerted its effects through the inhibition of the epidermal growth factor receptor (EGFR) EGFR-Akt-ERK signaling pathway, as evidenced by reduction of p-EGFR, p-Akt, and p-ERK in a western blot analysis in HCC827 cells. These findings suggest that lotusine exerts potent anti-cancer effects via a multifaceted mechanism, including inhibition of proliferation, apoptosis induction, and cell cycle arrest, predominantly mediated by EGFR suppression. This study highlights lotusine as a promising therapeutic candidate for the treatment of EGFR-mutant NSCLC and provides insights into its molecular mechanisms of action, paving the way for further preclinical and clinical evaluations.
Collapse
Affiliation(s)
- Yuanmin Lan
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Jing Sun
- Department Of Oncology, The Fifth People's Hospital Of Dalian, Dalian Liaoning, 116021, China
| | - Jiqing Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen and Longgang District People's Hospital of Shenzhen, Guangdong, Guangdong, 518172, China
| | - Xiaoying Chen
- Department of Respiratory and Critical Care Medicine, Lishui Second People's Hospital, Lishui Zhejiang, 323000, China.
| |
Collapse
|
4
|
Su YF, Tsai TH, Kuo KL, Wu CH, Su HY, Chang WC, Huang FL, Lieu AS, Kwan AL, Loh JK, Lin CL, Tsai CY. Mitochondrial dysfunction and cell death induced by Toona sinensis leaf extracts through MEK/ERK signaling in glioblastoma cells. PLoS One 2025; 20:e0320849. [PMID: 40343958 PMCID: PMC12063860 DOI: 10.1371/journal.pone.0320849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/26/2025] [Indexed: 05/11/2025] Open
Abstract
Toona sinensis, a kind of phytochemicals in traditional Chinese medicine widely used in South-East Asia, has been recognized for its anticancer properties, particularly against various types of cancer. We aimed to evaluate the effectiveness of T. sinensis leaf extracts (TSL) specifically for glioblastoma multiforme (GBM). Gallic acid was identified as the major active component in the aqueous extracts of TSL using the HPLC system. Furthermore, it has been shown to have the ability to penetrate the blood-brain barrier. Various concentrations of TSL (10, 20, 40, and 80 μg/mL) were applied and 80 μg/mL TSL treatment significantly inhibited cell growth, proliferation, and cytotoxicity in A172 and U251 GBM cells. Flow cytometry analysis revealed cell cycle arrest at the G2/M phase and increased apoptotic cells. Furthermore, we observed mitochondrial dysfunction characterized by elevated ROS levels and reduced ATP production due to the blockade of electron transport chain (ETC) complexes. TSL treatment regulated this ROS-induced mitochondrial dysfunction. Western blotting analysis showed upregulation of Bax and Puma, along with downregulation of Bcl-2. Additionally, TSL treatment induced the cleavage of caspase-3, caspase-9, and PARP, indicating activation of the mitochondria-mediated apoptosis pathway and caspase-dependent pathway in both GBM cell lines. To investigate the involvement of the MEK/ERK pathway in TSL-induced effects, we used U0126, an inhibitor of MEK/ERK kinase. The results demonstrated that TSL treatment suppressed MEK/ERK activation, inhibiting ROS-induced mitochondrial dysfunction and promoting apoptosis. This suggests a potential therapeutic strategy targeting the MEK/ERK pathway in GBM treatment. Overall, our findings indicate that TSL treatment exerts cytotoxic effects through ROS-mediated mitochondrial dysfunction and activation of apoptotic pathways via MEK/ERK pathway in GBM cells. These insights provide valuable knowledge for potential therapeutic applications of TSL in GBM treatment.
Collapse
Affiliation(s)
- Yu-Feng Su
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tai-Hsin Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Keng-Liang Kuo
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chieh-Hsin Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Yuan Su
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wen-Chang Chang
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Fu-Long Huang
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Joon-Khim Loh
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng Yu Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Barzegar-Fallah A, Ghaffari-Bohlouli P, Nadjafi S, Razmi A, Dehpour AR, Ghaffarian-Bahraman A, Alimoradi H, Shafiei M. Tropisetron attenuates high-glucose-induced vascular endothelial dysfunction via inhibition of calcineurin/NFAT signalling. Eur J Pharmacol 2025; 994:177389. [PMID: 39961489 DOI: 10.1016/j.ejphar.2025.177389] [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: 09/07/2024] [Revised: 02/01/2025] [Accepted: 02/13/2025] [Indexed: 03/15/2025]
Abstract
Vascular endothelial dysfunction (VED) is considered an important initiating factor in pathogenesis of diabetic vascular disease. In this process, oxidative insult, cellular hypertrophy, and activation of the calcineurin/nuclear factor of activated T-cell (NFAT) pathway play key roles. Herein, we investigated the effects of tropisetron (TRS), a calcineurin inhibitor, on high glucose (HG)-induced hypertrophy and apoptosis in human umbilical vein endothelial cells (HUVECs). To this end, HUVECs and chorioallantoic membranes (CAMs) were exposed to HG with or without TRS or cyclosporine A (CsA), and the effects of the treatments were evaluated on oxidative stress generation, cell number (proliferation and apoptosis), cell size (hypertrophy), and vessel formation. We also explored the possible role of calcineurin-NFAT signalling in the potential protective effects of TRS on hypertrophy and apoptosis associated with HG. The average size and protein content of the cells exposed to HG for 48h were significantly increased compared with normal glucose (NG). HG significantly increased apoptosis, altered the cell cycle, and elevated oxidative and nitrosative stress in HUVECs. Further, exposing cells to HG resulted in elevated calcineurin activity and NFATc1 translocation to the nuclei. HG also caused a significant decrease in the formation of new blood vessels in CAMs. Inhibition of calcineurin/NFAT pathway by TRS or CsA protected against these pathological changes. Our data demonstrated that inhibition of calcineurin/NFAT signalling by TRS, as a safe calcineurin inhibitor, may ameliorate HG-induced VED. Further in vivo and clinical studies are required to fully determine the protective effects of TRS against VED in diabetes.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; BioMatter Unit-Biomass Transformation Lab (BTL), École Interfacultaire de Bioingénieurs (EIB), Université Libre de Bruxelles, Brussels, Belgium
| | - Pejman Ghaffari-Bohlouli
- BioMatter Unit-Biomass Transformation Lab (BTL), École Interfacultaire de Bioingénieurs (EIB), Université Libre de Bruxelles, Brussels, Belgium
| | - Shabnam Nadjafi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Razmi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Ghaffarian-Bahraman
- Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Houman Alimoradi
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; BioMatter Unit-Biomass Transformation Lab (BTL), École Interfacultaire de Bioingénieurs (EIB), Université Libre de Bruxelles, Brussels, Belgium.
| | - Massoumeh Shafiei
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Eswaran S, Mascarenhas R, Kabekkodu SP. The ester derivative Palmitoylcarnitine abrogates cervical cancer cell survival by enhancing lipotoxicity and mitochondrial dysfunction. Cell Commun Signal 2025; 23:213. [PMID: 40319292 PMCID: PMC12048960 DOI: 10.1186/s12964-025-02218-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND In cervical cancer (CC), Double C2 Like Domain Beta (DOC2B) functions as a metastatic suppressor. The present study aims to determine whether ectopic expression of DOC2B causes global metabolomic changes in extracellular vesicles (EVs) and corresponds with its tumor suppressive properties. METHODS Using a retroviral method, we first ectopically expressed DOC2B in SiHa cells, which do not normally express DOC2B. RESULTS We observed that ectopically expressed DOC2B significantly altered the global metabolite profile of EVs. Metabolomics identified significant enrichment of palmitoylcarnitine (PC) in EVs upon ectopic expression of DOC2B. We identified that SiHa and HeLa cells exhibited greater cytotoxicity to PC than gingival fibroblast, HaCaT, Cal27, and MCF7. PC treatment reduced the growth, proliferation, and migration of SiHa and HeLa cells, via increasing apoptosis and decreasing S-Phase cells. PC treatment resulted in morphological alterations, decreased length and number of filopodia, and expression of proteins related to cell cycle progression, proliferation, and the epithelial-to-mesenchymal transition. Further, PC treatment caused mitochondrial morphological changes, increased mitochondrial membrane potential, and decreased mtDNA content. The decreased GSH activity, glucose consumption rate, and lactate production upon PC treatment suggest that PC can induce metabolic reprogramming in CC cells. Increased oxidative stress, calcium overload, lipid droplet accumulation, mitochondrial lipotoxicity, and mitophagy suggest that PC can cause mitochondrial dysfunction. N-acetyl cysteine (NAC) treatment reversed the cytotoxic effect of PC, via decreasing lipid peroxidation rate and increasing GSH activity. PC treatment enhanced the cytotoxic effect of cisplatin in CC. CONCLUSION DOC2B restoration or the use of PC may be employed as a novel therapeutic approach for CC.
Collapse
Affiliation(s)
- Sangavi Eswaran
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Roshan Mascarenhas
- Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK
- Newcastle University Medicine Malaysia (NUMed), 1, Jalan Sarjana 1, Kota Ilmu, Educity@Iskandar, Iskandar Puteri, Johor, 79200, Malaysia
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
7
|
Lima LEBD, Almeida MLGD, Gomes GS, do Nascimento PH, Silva CJOE, da Silva CRD, Tanaka YMR, Romão TP, de Lima TBS, de Araújo ES, de Araújo PLB, Cabral Filho PE, Holanda VN, de Oliveira RN, Figueiredo RCBQD. A phthalimide-triazole derivative obtained by click chemistry exhibits trypanocidal activity, induces autophagy and ameliorates Trypanosoma cruzi infection. Biomed Pharmacother 2025; 186:117963. [PMID: 40101588 DOI: 10.1016/j.biopha.2025.117963] [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: 01/09/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi, remains a leading cause of cardiomyopathy and heart failure in Latin America. Since the 1970s, benznidazole (BNZ) and nifurtimox (NFX) have been the only chemotherapeutic agents used to treat CD. However, their toxicity and low effectiveness in the chronic phase of the disease, make the development of more efficient chemotherapeutics imperative. Here, we investigated the effects of 1,2,3-triazole hybrids, synthesized via click chemistry, containing either phthalimide (FT1, FT2, FT3, FT4) or naphthoquinone (NT1) moieties on T. cruzi and their cytotoxicity on mammalian cells. NT1 and FT1 were the most effective against intracellular parasite with an IC50 = 31.1 and 189.2 µM, respectively. FT1-FT4 showed low cytotoxicity to mammalian cells (CC50 > 754 µM), while NT1 exhibited moderate toxicity (CC50 ≥ 96.1 µM). FT1 demonstrated the highest selectivity towards trypomastigotes and amastigotes with selectivity indexes (SeI) of 6.9 and 6.7, respectively. Ultrastructural analysis of trypomastigotes treated with FT1 revealed mitochondrial alterations, lipid accumulation and Golgi complex disorganization. FT1 also decreased the mitochondrial membrane potential, increased mitochondrial reactive oxygen species (ROS) production, and induced late apoptosis in trypomastigotes. In infected cardiac cells, FT1 treatment led to degradation of amastigotes and Golgi disruption. An increase in autophagosomes in treated host cells and their interaction with intracellular parasites suggest that FT1-induced host cell autophagy may play a role in mitigating the infection and protecting cardiac cells from its deleterious effects.
Collapse
Affiliation(s)
- Lucas Eduardo Bezerra de Lima
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Maria Letícia Gomes de Almeida
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Gleicyane Silva Gomes
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Pedro Henrique do Nascimento
- Departamento de Microbiologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Carla Jasmine Oliveira E Silva
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil
| | - Cecilãne Regina Dioclecia da Silva
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil
| | - Yuri Mouzinho Ramos Tanaka
- Departamento de Entomologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego, s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Tatiany Patrícia Romão
- Departamento de Entomologia, Instituto Aggeu Magalhães, FIOCRUZ-PE, Avenida Prof° Moraes Rego, s/n°- Campus da UFPE, Recife, PE 50670420, Brazil
| | - Thaíses Brunelle Santana de Lima
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco (UFPE), Av. Prof. Luiz Freire, 1000, Cidade Universitária, Recife, PE 50740-545, Brazil
| | - Elmo Silvano de Araújo
- Departamento de Energia Nuclear, Universidade Federal de Pernambuco (UFPE), Av. Prof. Luiz Freire, 1000, Cidade Universitária, Recife, PE 50740-545, Brazil
| | - Patricia Lopes Barros de Araújo
- Departamento de Engenharia Biomédica, Universidade Federal de Pernambuco (UFPE), Av. Jornalista Aníbal Fernandes, Cidade Universitária, Recife, PE 50740-560, Brazil
| | - Paulo Euzébio Cabral Filho
- Departamento de Biofísica e Radiobiologia, Universidade Federal de Pernambuco, Avenida Prof° Moraes Rego s/n°, Recife, PE 50670901, Brazil
| | - Vanderlan Nogueira Holanda
- Departmento de Biomedicina, Centro Universitário de Vitória de Santo Antão (UNIVISA), Vitória de Santo Antão, PE 55610-050, Brazil
| | - Ronaldo Nascimento de Oliveira
- Departamento de Química, Laboratório de Síntese de Compostos Bioativos, Universidade Federal Rural de Pernambuco, Rua Dom Manoel de Medeiros S/N, Recife, PE 52171900, Brazil.
| | | |
Collapse
|
8
|
Ganci D, D'Anna L, Abruscato G, Le Chevalier M, Quideau O, Cataldo S, Pettignano A, Rubino S, Chiarelli R, Barone G, Luparello C, Bonsignore R. Harnessing redox reactions for anticancer effects: A copper(II) Schiff base complex induces apoptosis in HepG2 liver cancer cells via ROS generation. J Inorg Biochem 2025; 270:112938. [PMID: 40344694 DOI: 10.1016/j.jinorgbio.2025.112938] [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: 12/02/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
This study uncovers the potential of a copper(II) Schiff base complex, CuL2+, to access the Cu(I) oxidation state and generate reactive oxygen species (ROS), highlighting its significance in eventual therapeutic applications. UV-vis absorption spectroscopy was used to follow the redox stability of the metal complex, also in the presence of reducing agents, such as ascorbic acid and glutathione, and of the copper(I) chelator, bathocuproine disulfonate. Utilizing human tumor cell lines HepG2 (hepatocarcinoma cells), we assessed its efficacy in reducing cell viability, increasing the sub-G0/G1 cell fraction, and initiating apoptotic pathways. Cell viability assays demonstrated a dose-dependent cytotoxicity with pronounced effects at sub-micromolar concentrations. Flow cytometry revealed significant ROS production, followed by mitochondrial membrane potential dissipation, and caspase activation, underscoring CuL2+'s mechanism of action. These findings position CuL2+ as a promising candidate for cancer therapy, providing insights into copper complexes' therapeutic application through oxidative stress and apoptosis modulation.
Collapse
Affiliation(s)
- Daniela Ganci
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Luisa D'Anna
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giulia Abruscato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | | | - Océane Quideau
- Institut National des Sciences Appliquées de Rouen, 76800 Saint-Étienne-du-Rouvray, France
| | - Salvatore Cataldo
- Dipartimento di Fisica e Chimica - Emilio Segrè, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Alberto Pettignano
- Dipartimento di Fisica e Chimica - Emilio Segrè, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Simona Rubino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Roberto Chiarelli
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Claudio Luparello
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy
| | - Riccardo Bonsignore
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, 90128 Palermo, Italy.
| |
Collapse
|
9
|
Li L, Che Y, Zhu L, Liu Z, Song Y, Wu Q, Chen M, Wang J. Calycosin, a Bioactive Isoflavone, Ameliorates Oxidative Stress and Inflammation in Lipopolysaccharide-Induced Intestinal Cell Damage Model via the Nrf2 and NF-κB Signaling Pathways. FASEB J 2025; 39:e70550. [PMID: 40244892 DOI: 10.1096/fj.202402387r] [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: 10/07/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Calycosin (CA), a bioactive isoflavone derived from Astragali Radix, has garnered interest in food sciences and pharmacology due to its potential antioxidant and anti-inflammatory properties. Our previous studies found that CA induced host defense peptide production in porcine IPEC-J2 cells and alleviated hydrogen peroxide-induced cellular oxidative damage. However, it is still unknown whether CA can protect against pathogenic microorganisms or toxins that cause intestinal cell damage. In this study, we aimed to investigate the protective effects of CA against LPS-induced intestinal cell damage. Using IPEC-J2 intestinal cells to develop a damage model, we found that LPS exposure caused significant morphological damage and apoptosis. However, pretreatment with CA effectively attenuated these adverse effects. Mechanistically, CA alleviated LPS-induced oxidative stress by reducing reactive oxygen species, malondialdehyde, 8-hydroxy-2'-deoxyguanine and carbonyl production, while enhancing antioxidant enzyme activities and gene expression. Furthermore, CA restored the LPS-induced decrease in nuclear erythroid 2-related factor 2 (Nrf2) expression, and Nrf2 silencing abolished the protective effects of CA, indicating that its action is mediated through the Nrf2 pathway. Additionally, CA suppressed LPS-induced inflammation by downregulating inflammation-related gene expression and inhibiting the nuclear factor (NF)-κB pathway. Collectively, our findings demonstrate that CA protects intestinal cells from LPS-induced damage by mitigating oxidative stress and inflammation via the Nrf2 and NF-κB pathways. These results suggest that CA has potential to be developed as a feed additive to prevent intestinal injury in animals, warranting further investigation in vivo.
Collapse
Affiliation(s)
- Lu Li
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yuyan Che
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Longlong Zhu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Zeqing Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Yanchen Song
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Qiong Wu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Wang
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
- Sino-US Joint Laboratory of Animal Science, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| |
Collapse
|
10
|
Song C, Zhao L, Deng J, Wang L, Mao M, Peng S, Tang W. E2F8-induced GRPEL2 promoted colorectal cancer progression via targeting TIGAR. J Transl Med 2025; 23:466. [PMID: 40269881 PMCID: PMC12020167 DOI: 10.1186/s12967-025-06451-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the leading cause for cancer mortality across the world. GRPEL2 is a critical regulator of mitochondria's function with an oncogenic role in different kinds of cancer. The exact function of GRPEL2 -driven mitochondrial regulation and CRC progression have not been elucidated. METHODS RNA-seq data from TCGA database was analyzed to identify biomarkers and therapeutic targets of CRC. The gene expression profile was validated by quantitative real-time PCR on 68 paired tumor and non-tumor samples from CRC patients. Tumorigenesis regulated by GRPEL2 was tested through EdU staining, Transwell assay, in vivo tumor growth and in vivo metastasis. The function of Mitochondria mediated by GRPEL2 was evaluated by transmission electron microscopy, DCFH-DA staining, mitochondrial membrane potential detection, and Calcein staining. LC-MS/MS screening and Co-IP were performed to discover protein partners of GRPEL2. E2F8-mediated transcriptional regulation of GRPEL2 was verified via Luciferase reporter and ChIP assays. RESULTS GRPEL2 was upregulated in CRC tumor tissues and cell lines. High expression of GRPEL2 was associated with poor prognosis of CRC and inhibition of GRPEL2 suppressed CRC proliferation and migration by inducing mitochondria injury. Meanwhile, TIGAR was shown to interact with GRPEL2 and overexpression of TIGAR rescued CRC progression in the presence of GRPEL2 inhibition. Moreover, E2F8 was the upstream regulator of GRPEL2, which positively induced GRPEL2 transcription and expression in CRC. CONCLUSION Our work illustrated the oncogenic role of GRPEL2 in CRC development and determined the molecular mechanism of E2F8/GRPEL2/TIGAR pathway. These findings will provide novel insights and promising therapeutic targets for CRC treatment in the future.
Collapse
Affiliation(s)
- Cheng Song
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Lei Zhao
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jing Deng
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Li Wang
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Min Mao
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Siyuan Peng
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Wei Tang
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Lushan Road, Yuelu District, Changsha, Hunan, China.
| |
Collapse
|
11
|
Li H, Tang S, Jia X, Zhu X, Cai L, Duan M, Wang S, Jiang H, Ji M, Wang S, Chen J. Combined toxicity evaluation of polystyrene nanoplastics and Nano-ZnO of distinctive morphology on human lung epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179097. [PMID: 40112547 DOI: 10.1016/j.scitotenv.2025.179097] [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: 09/23/2024] [Revised: 02/19/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
Despite increasing concerns on the co-exposure of nanoplastics (NPs) and heavy metals including zinc oxide nanoparticles (Nano-ZnO) in the public health, the systematic studies as well as available methodology of combined toxicity evaluation of Nano-ZnO/NPs are lacking. In this study, the single and combined toxicity of Nano-ZnO and polystyrene nanoplastics (PS-NPs) on human lung epithelial cells were evaluated by a combination of in vitro approach including real-time cell analysis (RTCA), cell counting kit-8 (CCK-8), oxidative stress, cell membrane integrity and apoptosis assay. RTCA was employed to dynamically monitor the effect of combined exposure of Nano-ZnO and PS-NPs on cell growth, in comparison with end-point CCK-8 assay. It was found that the cytotoxicity of different Nano-ZnO involved disintegration of cell membrane and causing oxidative stress and apoptosis while PS-NPs mainly induced oxidative stress and apoptosis. The proposed study not only pinpointed the distinctive interaction mode between Nano-ZnO and nanoplastics, but provided integrated approaches to environment and health risk assessment of co-exposed Nano-ZnO and nanoplastics.
Collapse
Affiliation(s)
- Henghui Li
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Simin Tang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaoyu Jia
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinyi Zhu
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ling Cai
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Mingxiu Duan
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Shaozhuo Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Huijun Jiang
- School of Pharmacy, Nanjing Medical University, 211166 Nanjing, China
| | - MingHui Ji
- School of Nursing, Nanjing Medical University, Nanjing 211166, China
| | - Shoulin Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Jin Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Province Engineering Research Center of Antibody Drug, Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
12
|
Wang Y, Yuan H, Fang R, Lu J, Duo J, Li G, Wang WJ. A new gold(I) phosphine complex induces apoptosis in prostate cancer cells by increasing reactive oxygen species. Mol Cell Biochem 2025; 480:2265-2276. [PMID: 38782835 DOI: 10.1007/s11010-024-05035-8] [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: 01/29/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Thioredoxin reductase (TrxR) is a pivotal regulator of redox homeostasis. It is frequently overexpressed in various cancer cells, including prostate cancer, making it a promising target for the development of anti-cancer drugs. In this study, we screened a series of newly designed complexes of gold(I) phosphine. Specifically, Compound 5 exhibited the highest cytotoxicity against prostate cancer cells and demonstrated stronger antitumor effects than commonly used drugs, such as cisplatin and auranofin. Importantly, our mechanistic study revealed that Compound 5 effectively inhibits the TrxR system in vitro. Additionally, Compound 5 promoted intracellular accumulation of reactive oxygen species (ROS), leading to mitochondrial dysfunction and irreversible apoptosis in prostate cancer cells. Our in vivo xenograft study further demonstrated that Compound 5 has excellent antitumor activity against prostate cancer cells, but does not cause severe side effects. These findings provide a promising lead Compound for the development of novel antitumor agents targeting prostate cancer and offer a valuable tool for investigating biological pathways involving TrxR and ROS modulation.
Collapse
Affiliation(s)
- Yuan Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Haokun Yuan
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiqin Fang
- The School of Life Science, University of Electronic Science and Technology of China, Chengdu, China
| | - Junzhu Lu
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiaqi Duo
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ge Li
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei-Jia Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, School of Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
13
|
Kuo JF, Hsiao YP, Wang YD, Weng HP, Wang CC. Fipronil Triggers Immunotoxicity Through Reactive Oxygen Species-Driven Mitochondrial Apoptosis in Thymocytes. TOXICS 2025; 13:204. [PMID: 40137531 PMCID: PMC11945543 DOI: 10.3390/toxics13030204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Fipronil (FPN), a widely used pesticide, is associated with significant immunotoxic effects, particularly impacting thymocyte survival and immune homeostasis. This study explores the mechanistic pathways underlying FPN-induced apoptosis and oxidative stress. Short-term FPN exposure (1-10 mg/kg) notably suppressed the expression of both anti-apoptotic (Bcl-2, Bcl-6, Mcl-1) and pro-apoptotic (Bnip3, Bim) genes in thymic tissues in vivo. Additionally, in isolated primary thymocytes, FPN directly decreased the expression of Bcl-2, Bcl-6, Mcl-1, and Bnip3 expression, coupled with a significant increase in pro-apoptotic Bim expression in a dose-dependent manner. FPN treatment directly led to elevated reactive oxygen species (ROS), lipid peroxidation, mitochondrial membrane depolarization, reduced cellular metabolic activity, and depleted intracellular calcium and glutathione (GSH) levels, indicating mitochondrial dysfunction and oxidative stress. Annexin V/PI staining confirmed that FPN induced late-stage apoptosis and necrosis in primary thymocytes. These findings elucidate the immunotoxic effects of FPN on thymocytes, highlighting its detrimental impact on immune system integrity, thymic development, and T cell maturation through oxidative damage and mitochondrial-mediated apoptosis.
Collapse
Affiliation(s)
- Jui-Fang Kuo
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (J.-F.K.); (Y.-D.W.)
| | - Yai-Ping Hsiao
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA;
| | - Yao-De Wang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (J.-F.K.); (Y.-D.W.)
| | - Hsin-Pei Weng
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (J.-F.K.); (Y.-D.W.)
| | - Chia-Chi Wang
- School of Veterinary Medicine, National Taiwan University, Taipei 106, Taiwan; (J.-F.K.); (Y.-D.W.)
| |
Collapse
|
14
|
Manmuan S, Sirirak T, Tubtimsri S, Petchsomrit A, Chuenbarn T. Phytochemical analysis, antioxidant activity, and cytotoxic effects of Caulerpa lentillifera extracts inducing cell apoptosis and sub-G/G0-G1 cell cycle arrest in KON oral cancer cells. BMC Complement Med Ther 2025; 25:101. [PMID: 40069755 PMCID: PMC11899751 DOI: 10.1186/s12906-025-04835-9] [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: 06/08/2024] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Marine algae have excellent phytoconstituents with notable biological activity and bioactive therapeutic benefits, but the anti-oral cancer activity of Caulerpa lentillifera (C. lentillifera) has not been widely studied. This study aimed to explore the anti-cancer properties of C. lentillifera to gain insights into possible treatment approaches. METHODS The three C. lentillifera extracts were prepared using the maceration method with methanol (CLM), ethanol (CLE), and acetone (CLA). The chemical composition of extracts of C. lentillifera was investigated. Its metabolite profiles were selectively further investigated using the LC-QTOF MS/MS technique and their antioxidative activity was evaluated. The cytotoxic effect on KON cells and MRC-5 cells was assessed using the MTT test. Morphological changes and apoptosis were examined through Hoechst 33,258 and AO double staining, while DAPI and FDA double labeling were used to observe the nucleus and cytoplasm. Using a flow cytometer, the percentage of cell cycle arrest was calculated and the fraction of cell death was examined. RESULTS The CLA exhibited higher quantities of TPC, TFC, chlorophyll a, and chlorophyll b compared to the CLM and CLE. The LC-QTOF MS/MS analysis revealed ten major phytochemicals in the CLA. The three C. lentillifera extracts exhibited antioxidative activity, with the CLE demonstrating significantly higher antioxidant activity compared to the CLM and CLA. In-vitro, the KON oral cancer cells exhibited sensitivity to CLA, CLE, and CLM in that order. The three extracts induced ROS-mediated cell death as well as disruption of mitochondrial membrane potential, with concentrations at IC40, IC60, and IC80 leading to apoptosis within 24 h. Furthermore, the cell cycle of KON cells was blocked in sub-G and G0-G1 by all three extracts. Notably, the extracts significantly impeded colony growth, migration, and invasion. The increase in cellular uptake was measured using the TEER test. CONCLUSION The findings showed that C. lentillifera has several functional metabolites, antioxidative activity, and strong anti-tumor properties. According to these results, C. lentillifera extracts may be utilized to treat oral cancer.
Collapse
Affiliation(s)
- Suwisit Manmuan
- Division of Pharmacology and Biopharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand.
| | - Thanchanok Sirirak
- Division of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Sukannika Tubtimsri
- Division of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Arpa Petchsomrit
- Division of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Tiraniti Chuenbarn
- Division of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
15
|
Jhetam Z, Martins-Furness C, Slabber C, Munro OQ, Nel M, Harmse L. Copper complexes induce haem oxygenase-1 (HMOX1) and cause apoptotic cell death in pancreatic cancer cells. J Inorg Biochem 2025; 264:112815. [PMID: 39740375 DOI: 10.1016/j.jinorgbio.2024.112815] [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: 07/27/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/02/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common pancreatic malignancy, has a dismal 5-year survival rate, making palliative chemotherapy the only treatment option. Targeted therapy has limited efficacy in PDAC, underscoring the need for novel therapeutic approaches. The inducible stress-response protein, haem oxygenase-1 (HMOX1), has been implicated in treatment failure in PDAC. Copper coordination complexes have shown promise as anticancer agents against various cancers, and are associated with apoptotic cell death. The different ligands to which copper is complexed, determine the specificity and efficacy of each complex. Three different classes of copper complexes were evaluated for anti-cancer activity against AsPC-1 and MIA PaCa-2 pancreatic cancer cell lines. A copper-phenanthroline-theophylline complex (CuPhTh2), a copper-8-aminoquinoline-naphthyl complex (Cu8AqN), and two copper-aromatic-isoindoline complexes (CuAIsI) were effective inhibitors of cell proliferation with clinically relevant IC50 values below 5 μM. The copper complexes caused reactive oxygen species (ROS) formation, promoted annexin-V binding, disrupted the mitochondrial membrane potential (MMP) and activated caspase-9 and caspase-3/7, confirming apoptotic cell death. Expression of nuclear HMOX1 was increased in both cell lines, with the CuPhTh2 complex being the most active. Inhibition of HMOX1 activity significantly decreased the IC50 values of these copper complexes suggesting that HMOX1 inhibition may alter treatment outcomes in PDAC.
Collapse
Affiliation(s)
- Zakeeya Jhetam
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Carla Martins-Furness
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Cathy Slabber
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, 1 Jan Smut Ave, Braamfontein, Johannesburg 2017, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, UK
| | - Marietha Nel
- Dept of Surgery, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa
| | - Leonie Harmse
- Division of Pharmacology, Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
16
|
Ahn CR, Baek SH. Synergistic effects of 6-shogaol and hyperthermia on ACHN renal cancer cells: modulation of ROS and heat shock pro-teins in cancer therapy. Front Pharmacol 2025; 16:1522285. [PMID: 40051570 PMCID: PMC11882530 DOI: 10.3389/fphar.2025.1522285] [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: 11/04/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Renal cancer is known for its aggressive progression and resistance to standard treatments, underscoring the need for novel therapeutic strategies. This study explores the potential of combining 6-shogaol (6-SHO), a bioactive compound derived from ginger (Zingiber officinale), with hyperthermia to enhance anticancer efficacy in ACHN renal cancer cells. Methods ACHN cells were treated with 6-SHO and exposed to hyperthermic conditions. We evaluated the combined effects on apoptosis, cell cycle arrest, and cell proliferation, as well as the role of reactive oxygen species (ROS) and heat shock proteins (HSPs) in mediating these responses. Results The combination of 6-SHO and hyperthermia significantly increased apoptosis, induced G2/M phase cell cycle arrest, and reduced cell proliferation more effectively than either treatment alone. ROS played a critical role in these effects, with modulation of HSPs and heat shock factor 1 (HSF1) further disrupting cancer cell survival mechanisms. Discussion These findings highlight the synergistic potential of 6-SHO and hyperthermia as a novel therapeutic approach in renal cancer treatment, supporting the need for further research and clinical evaluation.
Collapse
Affiliation(s)
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
17
|
Lin F, Recchia MJJ, Clark TN, Kennelly EJ, Linington RG, Long C. Metabolite profiling and characterization of potential anticancer constituents from Garcinia subfalcata using UPLC-IMS-QTOF-MS. Food Chem 2025; 465:141900. [PMID: 39531968 DOI: 10.1016/j.foodchem.2024.141900] [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: 05/24/2024] [Revised: 10/19/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Garcinia subfalcata, an edible species endemic to China, has limited research on its chemical composition and biological effects. This study aimed to analyze metabolites in different plant parts and identify potential anticancer constituents. Using UPLC-IMS-QTOF-MS-based metabolomics, a total of 124 compounds were identified, with xanthones, flavonoids and phloroglucinols being the predominant compounds. PCA and PLS-DA analyses revealed significant metabolite differences among plant parts, identifying 28 differential metabolites, including bronianone and (±)-fukugiside. Antiproliferative assays showed varying bioactivities, with bark exhibiting the highest cytotoxicity against A549, HeLa and HGC-27 cells (IC50: 2.72-5.71 μg/mL). Mechanism studies indicated that the bark inhibited cell proliferation by inducing apoptosis and disrupting mitochondrial membrane potential. S-plot models revealed 23 potential anticancer constituents, including (-)-epicatechin and 1,7-dihydroxyxanthone. These findings highlight G. subfalcata's potential as a source of functional food supplements and medicinal agents and indicate the efficacy of UPLC-IMS-QTOF-MS-based metabolomics in exploring bioactive components within Garcinia.
Collapse
Affiliation(s)
- Fengke Lin
- College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, PR China; School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, PR China
| | | | - Trevor N Clark
- Department of Chemistry, Simon Fraser University, Burnaby V5A 1S6, Canada; Department of Chemistry, University of Prince Edward Island, Charlottetown C1A 4P3, Canada
| | - Edward J Kennelly
- Department of Biological Sciences, Lehman College, City University of New York, New York 10468, United States; PhD Program in Biology, The Graduate Center, City University of New York, New York 10016, United States
| | - Roger G Linington
- Department of Chemistry, Simon Fraser University, Burnaby V5A 1S6, Canada.
| | - Chunlin Long
- College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, PR China; Key Laboratory of Ecology and Environment in Minority Areas (Minzu University of China), National Ethnic Affairs Commission of China, Beijing 100081, PR China; Key Laboratory of Ethnomedicine, Minzu University of China, Ministry of Education, Beijing 100081, PR China; Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission of China, Beijing 100081, PR China.
| |
Collapse
|
18
|
Jurčacková Z, Hrčková G, Mudroňová D, Matiašová AA, Biedermann D. Flavonolignans silybin, silychristin and 2,3-dehydrosilybin showed differential cytoprotective, antioxidant and anti-apoptotic effects on splenocytes from Balb/c mice. Sci Rep 2025; 15:5631. [PMID: 39955331 PMCID: PMC11830019 DOI: 10.1038/s41598-025-89824-1] [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: 07/10/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025] Open
Abstract
Silymarin is an extract obtained from the seeds of milk thistle (Sylibum marianum L., Asteraceae) and contains several structurally related flavonolignans and a small family of flavonoids. Mouse spleen cells represent highly sensitive primary cells suitable for studying the pharmacological potential and biofunctional properties of natural substances. Cultivation of splenocytes for 24 h under standard culture conditions (humidity, 37 °C, 5% CO2, atmospheric oxygen) resulted in decreased viability of splenocytes compared to intact cells. A cytoprotective effect of silybin (SB), silychristin (SCH) and 2,3-dehydrosilybin (DHSB) was observed at concentrations as low as 5 µmol/ml. At 50 µmol/ml, these substances restored and/or stimulated viability and mitochondrial membrane potential and had anti-apoptotic effect in the order SB > DHSB > SCH. The substances demonstrated a concentration-dependent activity in restoring the redox balance based on the changes in the concentration of reactive oxygen species (ROS), hydrogen peroxide (H2O2) and nitric oxide. This was in the order DHSB > SCH > SB, which correlated with the suppressed expression of nuclear factor erythroid 2-related factor 2 (Nrf2), catalase and glutathione peroxidase. The strong stimulation of the superoxide dismutase 1 gene converting ROS to H2O2 points to its dominant role in the maintaining redox homeostasis in splenocytes, which was disrupted by oxidative stress due to non-physiological culture conditions. Our study showed significant differences in the cytoprotective, antioxidant and anti-apoptotic activities of SB, SCH, and DHSB on splenocytes exposed to mild and AAPH-induced oxidative stress.
Collapse
Affiliation(s)
- Zuzana Jurčacková
- Institute of Parasitology, Slovak Academy of Sciences, Hlinkova 3, 04001, Kosice, Slovakia
| | - Gabriela Hrčková
- Institute of Parasitology, Slovak Academy of Sciences, Hlinkova 3, 04001, Kosice, Slovakia.
| | - Dagmar Mudroňová
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | - Anna Alexovič Matiašová
- Institute of Biology and Ecology, Faculty of Science, P. J. Šafárik University, Kosice, Slovakia
| | - David Biedermann
- Institute of Microbiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
19
|
Qudus MS, Afaq U, Liu S, Wu K, Yu C, Tian M, Wu J. SARS-CoV-2-ORF-3a Mediates Apoptosis Through Mitochondrial Dysfunction Modulated by the K + Ion Channel. Int J Mol Sci 2025; 26:1575. [PMID: 40004042 PMCID: PMC11855091 DOI: 10.3390/ijms26041575] [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: 12/31/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) causes pulmonary edema, which disrupts the lung alveoli-capillary barrier and leads to pulmonary cell apoptosis, the main cause of death. However, the molecular mechanism behind SARS-CoV-2's apoptotic activity remains unknown. Here, we revealed that SARS-CoV-2-ORF-3a mediates the pulmonary pathology associated with SARS-CoV-2, which is demonstrated by the fact that it causes lung tissue damage. The in vitro results showed that SARS-CoV-2-ORF-3a triggers cell death via the disruption of mitochondrial homeostasis, which is modulated through the regulation of Mitochondrial ATP-sensitive Potassium Channel (MitoKATP). The addition of exogenous Potassium (K+) in the form of potassium chloride (KCl) attenuated mitochondrial apoptosis along with the inflammatory interferon response (IFN-β) triggered by SARS-ORF-3a. The addition of exogenous K+ strongly suggests that dysregulation of K+ ion channel function is the central mechanism underlying the mitochondrial dysfunction and stress response induced by SARS-CoV-2-ORF-3a. Our results designate that targeting the potassium channel or its interactions with ORF-3a may represent a promising therapeutic strategy to mitigate the damaging effects of infection with SARS-CoV-2.
Collapse
Affiliation(s)
- Muhammad Suhaib Qudus
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Uzair Afaq
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Chen Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China or (M.S.Q.); (U.A.); (S.L.); (K.W.); (J.W.)
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
20
|
Xuan Y, Yan Y, Wei X, Wang S, Zhang J, Tang Y, Li S. Positively-charged, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes functionalized with ethacrynic acid: Synthesis, characterizaion, and antitumor effect. J Inorg Biochem 2025; 263:112778. [PMID: 39615317 DOI: 10.1016/j.jinorgbio.2024.112778] [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: 09/18/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
A new family of ethacrynic acid-functionalized, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes (4a-4e) have been designed, synthesis and fully characterized by 1H and 13C NMR, ESI-MS, elemental analysis, and melting point tests. The molecular structure of 3a, one of the precursor complexes, has been determined by single-crystal X-ray diffraction. The cytotoxicity of the obtained complexes toward human cancer cell lines such as HeLa, MGC803, A549, MDA-MB-231, and MCF-7 cells have been investigated by MTT assay. Whereas complexes 4d and 4e showed significantly higher cytotoxicity than cisplatin (the positive control group) and complexes 3a-3e. Moreover, complexes 4d and 4e exhibited a certain selectivity (selectivity index: 7.33 and 7.57) toward MCF-7 cells over MCF-10a normal cells. Glutathione S-transferases (GSTs) activity assay indicate that complexes 4d and 4e exhibited higher GST inhibitory activity than ethacrynic acid (EA, the best characterized GST inhibitor), consistent with their higher cytotoxicity. Further mechanistic studies showed that 4e-induced cell apoptosis may be aroused by the production of ROS, the loss of mitochondrial membrane potential and G2/M phase cell arrest in MCF-7 cells. In addition, the in vivo antitumor effect study on the xenograft mouse models of MCF-7 cells reveal that complex 4e significantly inhibited tumor growth with a higher inhibition efficiency of 68.80 %, in comparison with the groups treated with cisplatin (59.25 %). These results highlight the strong possibility to develop positively-charged, chalcone-hydroxypyrone hybrid ruthenium(II)-arene complexes funcionalized with GST inhibitor as promising anticancer agents.
Collapse
Affiliation(s)
- Yuxin Xuan
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Yuxi Yan
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Xiaonan Wei
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Shuxiang Wang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Jinchao Zhang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China
| | - Yonghe Tang
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China.
| | - Shenghui Li
- Key Laboratory of Chemical Biology of Hebei Province; Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education; State Key Laboratory of New Pharmaceutical Preparations and Excipients, College of Chemistry & Materials Science, Hebei University, Baoding 071002, China.
| |
Collapse
|
21
|
Gao S, Li J, Wang W, Wang Y, Shan Y, Tan H. Rabdosia rubescens (Hemsl.) H. Hara: A potent anti-tumor herbal remedy - Botany, phytochemistry, and clinical applications and insights. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119200. [PMID: 39631716 DOI: 10.1016/j.jep.2024.119200] [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: 09/14/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese herbal medicine has unique advantages as anti-cancer drugs and adjuvant therapies. Rabdosia rubescens (Hemsl.) H. Hara (R. rubescens) is a traditional medicinal plant known for its anti-inflammatory, antioxidant, antibacterial, anti-angiogenic and antitumor properties. The antitumor activity of R. rubescens is widely recognized among the folk communities in Henan Province, China. AIM OF THE STUDY This study reviews the botany, ethnopharmacology, phytochemistry, anti-tumor active ingredients, mechanisms, and clinical applications of R. rubescens, aiming to provide a comprehensive understanding for its use as an anti-cancer drug and adjuvant therapy. MATERIALS AND METHODS We systematically searched the literature in PubMed, Web of Science, and CNKI using the following keywords: "Rabdosia rubescens", "Isodon rubescens", "traditional application", "anti-tumor", "phytochemistry", "anti-tumor active compounds", "oridonin" and "clinical application". The search covered publications from 1997 to 2024. Inclusion criteria included original studies or reviews focusing on the anti-tumor properties of R. rubescens or its active components. Exclusion criteria included studies related to non-R. rubescens applications. RESULTS R. rubescens is a perennial herbaceous plant in the family Lamiaceae, mainly found in central and southern China. Historically, it has been used to treat conditions such as sore throat, cough, and excess phlegm. The plant contains various compounds, including diterpenes, triterpenes, steroids, flavonoids, phenolic acids, essential oils, amino acids, alkaloids, and polysaccharides, with diterpenes, triterpenes, flavonoids, and phenolic acids being the most active. This review identifies 50 compounds with anti-tumor properties, comprising 34 diterpenes, 2 triterpenes, 7 flavonoids, and 7 phenolic acids. Notably, besides oridonin and ponicidin, the ent-kaurane diterpenoids (20S)-11β,14β,20-trihydroxy-7α,20-epoxy-ent-kaur-16-en15-one and (20S)-11β,14β-dihydroxy-20-ethoxy7α,20-epoxy-ent-kaur-16-en-15-one demonstrate significant anti-tumor activity, attributed to their carbonyl group at C-15, hydroxyl group at C-1, and OEt group at C-20. Mechanistically, R. rubescens combats tumors by blocking the tumor cell cycle, promoting apoptosis, inhibiting cell migration and angiogenesis, inducing ferroptosis, reversing drug resistance, and enhancing radiosensitivity in tumor cells. Clinically, R. rubescens is available in various forms, including tablets, drops, syrups, capsules, and lozenges, and is primarily used for tonsillitis, pharyngitis, and stomatitis. According to the 2020 edition of the Pharmacopoeia of China, R. rubescens tablets are recognized as an adjuvant therapy for cancer. Clinical studies indicate that R. rubescens syrup, tablets, and thermal therapy can enhance cancer patient survival rates and lower tumor recurrence rates. CONCLUSIONS Given its traditional and modern uses, active anti-tumor components, and mechanisms, R. rubescens is a promising resource in traditional Chinese medicine for anti-tumor therapy. To realize its full potential, future research should explore additional active anti-tumor compounds beyond oridonin and ponicidin. For these key components, studies should focus on structural modifications to identify new active molecules and essential anti-tumor structures. Clinically, it is important to investigate how R. rubescens interacts with other Chinese herbs in anti-tumor formulations to enhance treatment efficacy and guide appropriate clinical use. Furthermore, future studies should undergo ethical review and include larger-scale randomized controlled trials to validate the efficacy of R. rubescens in treating tumors, thereby promoting its role as an anti-tumor traditional Chinese medicine.
Collapse
Affiliation(s)
- Shiyong Gao
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Jianwen Li
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Weiya Wang
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Yue Wang
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Yanmin Shan
- Drug Engineering and Technology Research Center, Harbin University of Commerce, Harbin, 150076, Heilongjiang, China; Heilongjiang Provincial Key Laboratory of Tumor Prevention and Antitumor Drugs, Harbin, 150076, Heilongjiang, China
| | - Huixin Tan
- Department of Pharmacy, Fourth Affiliated Hospital of Harbin Medicine University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
22
|
Ghorpade M, Rajput D, Mahalingam P, Kanvah S. Live cell imaging of lipid droplets: fluorescent chalcones as probes for lipophagy and lipid-mitochondria interactions. J Mater Chem B 2025; 13:1338-1349. [PMID: 39660366 DOI: 10.1039/d4tb01871k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Lipid droplets are crucial organelles involved in cellular energy storage and metabolism, which is key in maintaining energy homeostasis through lipophagy. In this work, we successfully synthesized donor-acceptor chalcone derivatives (M1-M3) with improved photophysical characteristics, such as significant Stokes shifts and strong emission features. DFT and TDDFT calculations have been employed to evaluate the structure-property relationship of the chalcone derivatives. The molecules show excellent selectivity in staining lipid droplets in COS-7 cells and other cell lines. The molecule M1 was also further utilized to monitor verapamil-induced lipophagy. Using M1, we also demonstrate the link between lipid droplets and mitochondria during stress, emphasizing the significance of lipophagy in cellular energy balance and metabolism. These results not only shed light on the lipid metabolism but also have profound implications for researching and potentially treating metabolic diseases, underscoring the importance of our work in the field.
Collapse
Affiliation(s)
- Mohini Ghorpade
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, India.
| | - Deeksha Rajput
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, India.
| | - Paramasivam Mahalingam
- School of Chemistry and Biochemistry and School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Sriram Kanvah
- Department of Chemistry, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382055, India.
| |
Collapse
|
23
|
Zhang L, Ding F, Ren Z, Cheng W, Dai H, Liang Q, Kong F, Xu W, Wang M, Zhang Y, Tao Q. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TNNI3 c.235C > T variant. Int J Cardiol 2025; 419:132627. [PMID: 39426416 DOI: 10.1016/j.ijcard.2024.132627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is typically manifested as a hereditary disorder, with 30 %-60 % of cases linked to cardiac sarcomere gene mutations. Despite numerous identified TNNI3 mutations associated with HCM, their severity, prevalence, and disease progression vary. The link between TNNI3 variants and phenotypes remains largely unexplored. This study aims to elucidate the impact of the TNNI3 c.235C > T mutation on HCM through clinical research and cell experiments and to explore its mechanism in HCM development. METHODS We screened an HCM family for pathogenic gene mutations using gene sequencing. The proband and family members were assessed through electrocardiography, echocardiography, and cardiac MRI, and a pedigree map was created for disease prediction analysis. Mutant plasmids were constructed with the TNNI3 c.235C > T mutation and transfected into the AC16 human cardiomyocyte cell line to investigate the mutation's effects. RESULTS The TNNI3 c.235C > T mutation was identified as the disease-causing variant in the family. This mutation led to the upregulation of hypertrophy-associated genes ANP, BNP, and MYH7, increased cardiomyocyte size, and activation of the ERK signaling pathway. Further investigations revealed that the TNNI3 c.235C > T mutation impaired mitochondrial function, disrupted cardiomyocyte metabolism, and increased cellular autophagy and apoptosis. CONCLUSIONS The TNNI3 c.235C > T gene mutation may be a pathogenic factor for HCM, showing heterogeneous features and clinical phenotypes. This mutation induces myocardial hypertrophy, activates the ERK signaling pathway, and exacerbates mitochondrial dysfunction, apoptosis, and autophagy in cardiomyocytes. These findings provide insights into the mechanism of HCM caused by gene mutations and may inform HCM treatment strategies.
Collapse
Affiliation(s)
- Lai Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fengzhi Ding
- Department of Physiology, Wannan Medical College, Wuhu, Anhui, 241000, China
| | - Zhongyuan Ren
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Weili Cheng
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - He Dai
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Qing Liang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fanling Kong
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Wenjing Xu
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Minghui Wang
- College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yuqing Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| | - Qin Tao
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
24
|
Ding M, Wang W, Huo K, Song Y, Chen X, Xiang Z, Chen P, Liu L. The Role of lncRNA FEZF1-AS1 in Colorectal Cancer Progression Via the P53 Signaling Pathway. DNA Cell Biol 2025; 44:32-45. [PMID: 39503758 DOI: 10.1089/dna.2024.0184] [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] [Indexed: 01/30/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical regulators in the development of colorectal cancer (CRC). Previous studies indicate that lncRNA FEZF1-AS1 is highly expressed in CRC, but its role in modulating CRC via the P53 signaling pathway remains unclear. In this study, we found that FEZF1-AS1 promotes the growth of the CRC cell line (HCT116) and drives epithelial-mesenchymal transition (EMT) through the P53 signaling pathway. Our data showed that FEZF1-AS1 expression is significantly upregulated in HCT116, and elevated levels of FEZF1-AS1 are associated with poor prognosis in patients with CRC. In addition, the knockdown of FEZF1-AS1 markedly inhibited the proliferation of HCT116 by inducing cell cycle arrest. Knockdown of FEZF1-AS1 depletion also led to apoptosis in CRC cells by suppressing the P53 signaling pathway and EMT, thereby reducing their viability, proliferation, migration, and invasion. In summary, this study confirmed that FEZF1-AS1 regulates the growth of junction HCT116 through P53 signaling pathway and inhibiting EMT, providing new insights for the potential therapeutic strategies against CRC.
Collapse
Affiliation(s)
- Minglu Ding
- Mudanjiang Medical University, Mudanjiang, China
| | - Wanyao Wang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Keyuan Huo
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Yidan Song
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Xiaojie Chen
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Zihan Xiang
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Peijian Chen
- College of Life Sciences, Mudanjiang Medical University, Mudanjiang, China
| | - Lantao Liu
- School of Basic Medicine, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
25
|
Shmagel K, Saidakova E, Korolevskaya L, Vlasova V, Younes SA. Activated/Cycling Treg Deficiency and Mitochondrial Alterations in Immunological Non-Responders to Antiretroviral Therapy. FRONT BIOSCI-LANDMRK 2024; 29:429. [PMID: 39735996 DOI: 10.31083/j.fbl2912429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, but their dynamics are altered in a subset of people living with Human Immunodeficiency Virus (HIV) known as immunological non-responders (INRs). INRs fail to reconstitute CD4+ T-cell counts despite viral suppression. This study aimed to examine Treg dysregulation in INRs, comparing them to immunological responders (IRs) and healthy controls (HCs). METHODS The study included 40 INRs, 42 IRs, and 23 HCs. Peripheral blood mononuclear cells were isolated and analyzed by flow cytometry. Conventional CD4+ T-cells (Tconvs) were identified as CD25-/loFOXP3- cells, while Tregs were identified as CD25+CD127loFOXP3+ CD4+ T-cells. Cells were further divided into naive, central memory, effector memory, and effector memory cells re-expressing CD45RA (TEMRA) subsets. Activated/cycling cells were identified as CD71+ and quiescent cells were CD71-. Mitochondrial mass and transmembrane potential were measured using MitoTracker Green and MitoTracker Orange dyes, respectively. Statistical comparisons were made using the Kruskal-Wallis test with Dunn's post-hoc analysis and Mann-Whitney U-test. RESULTS INRs exhibited the highest frequencies of activated/cycling CD4+ T-cells. The proportion of activated/cycling cells was higher in Tregs compared to Tconvs in all groups. Cycling rates of Tregs and Tconvs were correlated, suggesting Tregs help control Tconv proliferation. Despite high overall Treg frequencies in INRs, they showed a Treg deficiency in activated/cycling CD4+ T-cells, specifically in naive and central memory subsets, causing an imbalance in the Tconv/Treg ratio. This deficiency was hidden by increased Treg frequencies in quiescent effector memory CD4+ T-cells. Activated/cycling naive and memory Tregs from INRs had normal forkhead box P3 (FOXP3) and CD25 expression, but activated/cycling memory Tregs showed decreased ability to regulate mitochondrial transmembrane potential, indicating impaired mitochondrial fitness. These mitochondrial abnormalities were similar to those observed in memory conventional T-cells. CONCLUSIONS The complex Treg dysregulation in immunological non-responders involves quantitative and functional alterations, including a Treg deficiency within activated/cycling naive and central memory CD4+ T-cells, impaired mitochondrial fitness of activated/cycling memory Tregs, and functional disorders of the parent conventional T-lymphocytes. These findings underscore the need for a nuanced understanding of Treg dynamics in suboptimal CD4+ T-cell reconstitution during HIV-infection.
Collapse
Affiliation(s)
- Konstantin Shmagel
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm Federal Research Center UB RAS, 614081 Perm, Russian Federation
| | - Evgeniya Saidakova
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm Federal Research Center UB RAS, 614081 Perm, Russian Federation
- Biological Faculty, Perm State University, 614000 Perm, Russian Federation
| | - Larisa Korolevskaya
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm Federal Research Center UB RAS, 614081 Perm, Russian Federation
| | - Violetta Vlasova
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm Federal Research Center UB RAS, 614081 Perm, Russian Federation
| | - Souheil-Antoine Younes
- Pathology Advanced Translational Research Unit, Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
26
|
Lu Y, Zhang Y, Jin Z, Cui S, Wu L, He Y. Chiral Amino Acids Mediate Mitochondria-Dependent Apoptosis of Human Proximal Tubular Epithelial Cells Under Oxidative Stress. Int J Mol Sci 2024; 25:13439. [PMID: 39769204 PMCID: PMC11677210 DOI: 10.3390/ijms252413439] [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: 11/16/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Amino acids are the basic structural units of life, and their intake levels affect disease and health. In the case of renal disease, alterations in amino acid metabolism can be used not only as a clinical indicator of renal disease but also as a therapeutic strategy. However, the biological roles and molecular mechanisms of natural chiral amino acids in human proximal tubular epithelial cells (HK-2) remain unclear. In this study, cell viability assays revealed that chiral acidic amino acids (Glu and Asp) and aromatic amino acids (Trp and Phe) inhibited cell growth. The molecular mechanisms indicated that cell growth was closely related to ROS levels. Specifically, chiral Glu, Asp, Trp, and Phe induced oxidative stress and mitochondria-dependent apoptosis in HK-2 cells. This was manifested by elevated levels of intracellular ROS, 8-OHdG, and MDA, increased activities of antioxidant enzymes CAT, SOD, and GPx, decreased mitochondrial membrane potential, increased cytoplasmic Ca2+ concentration, and cell acidification. The expression levels of apoptosis-related molecules Caspase-9, Caspase-3, Cyt-C, and Bax were increased, and the expression level of anti-apoptotic molecule Bcl-2 was decreased. Moreover, L-Glu, D-Asp, L-Trp, and D-Phe exhibited a more pronounced inhibition of cell growth and elicited more substantial alterations in gene expression compared to the other configurations.
Collapse
Affiliation(s)
- Ying Lu
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (Y.L.); (Z.J.); (S.C.)
| | - Yang Zhang
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China;
| | - Zhaoyang Jin
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (Y.L.); (Z.J.); (S.C.)
| | - Shuaishuai Cui
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (Y.L.); (Z.J.); (S.C.)
| | - Li Wu
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (Y.L.); (Z.J.); (S.C.)
| | - Yujian He
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (Y.L.); (Z.J.); (S.C.)
- School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China;
| |
Collapse
|
27
|
Mondal R, Keerthana M, Pandurangan N, Shanmugaraju S. Zn(II)-Curcumin Complexes-Based Anticancer Agents. ChemMedChem 2024; 19:e202400558. [PMID: 39225342 DOI: 10.1002/cmdc.202400558] [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: 07/22/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/04/2024]
Abstract
There is a great deal of research interest in the design of alternative metallodrugs to Pt(II)-derivatives for cancer treatment. The low solubility of such drugs in biological mediums leading to poor bioavailability is the major hurdle of several metal-based anticancer agents. These issues have recently been addressed by designing bio-active ligands based on metal-containing anticancer agents. Conjugating with bioactive ligands has significantly improved the bioavailability of the metallodrugs and their cancer cell targeting ability. One such naturally available bioactive ligand is curcumin. Until recently, several curcumin-based anticancer metallodrugs have been developed and successfully demonstrated for their anticancer studies. In this article, we aim to highlight, the synthesis, structure, and anticancer properties of various Zn(II)-curcumin-based coordination complexes. The effect of introducing different functional groups, targeting ligands, and photo-active ligands on the anticancer potential of such complexes has been mentioned in detail. The current status and future perspective on curcumin-based metallodrugs for cancer treatment have also been stated.
Collapse
Affiliation(s)
- Rajdeep Mondal
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678557, India
| | - Muthukumar Keerthana
- Department of Chemistry, Indian Institute of Technology Palakkad, Palakkad, Kerala, 678557, India
| | - Nanjan Pandurangan
- Department of Chemistry, Amrita School of Physical Sciences, Amrita Vishwa Vidyapeetham, Coimbatore, Tamil Nadu, 641112, India
| | | |
Collapse
|
28
|
Quintal Bojórquez NDC, Segura Campos MR. Novel peptides derived from S. hispanica seeds induce selective cytotoxicity on human cancer cells. Food Chem 2024; 460:140470. [PMID: 39032303 DOI: 10.1016/j.foodchem.2024.140470] [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: 04/01/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Cancer prevails as one of the major health concerns worldwide due to the consistent rise in incidence and lack of effective therapies. Previous studies identified the peptides KLKKNL, MLKSKR, and KKYRVF from Salvia hispanica seeds and stated their selective anticancer activity. Thus, this study aimed to determine the cell death pathway induced by these peptides on five cancer cell lines (MCF-7, Caco2, HepG2, DU145, and HeLa). Based on the results of this work, it is possible to suggest that KLKKNL primarily induces selective cancer cell death through the apoptotic pathway in the Caco2 and HeLa lines. On the other hand, the peptide KKYRVF reported the highest statistical (p < 0.05) selective cytotoxic effect on the MCF-7, Caco2, HepG2, and DU145 cancer cell lines by induction of the necrotic pathway. These findings offer some understanding of the selective anticancer effect of KLKKNL, MLKSKR, and KKYRVF.
Collapse
Affiliation(s)
- Nidia Del Carmen Quintal Bojórquez
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico
| | - Maira Rubi Segura Campos
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico.
| |
Collapse
|
29
|
Gao L, Dalapati R, Gao B, Huang X, Zhao D, Wang F, Zang L. Mitochondrial STED Imaging and Membrane Potential Monitoring with a Cationic Molecular Probe. SMALL METHODS 2024; 8:e2400525. [PMID: 39268793 DOI: 10.1002/smtd.202400525] [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: 04/12/2024] [Revised: 09/05/2024] [Indexed: 09/15/2024]
Abstract
Mitochondria are essential organelles that not only undergo dynamic morphological changes but also exhibit functional activities such as mitochondrial membrane potential (MMP). While super-resolution techniques such as stimulated emission depletion (STED) nanoscopy can visualize the ultrastructure of mitochondria and the MMP probe can monitor mitochondria function, few dyes meet both demands. Here, a small molecule (MitoPDI-90) based on perylene diimide with cationic groups is reported and used for mitochondrial STED imaging and MMP indication. Characterized by excellent photostability, biocompatibility, and high quantum yield, MitoPDI-90 exhibits STED imaging compatibility, facilitating visualization of mitochondrial cristae and time-lapse imaging of highly dynamic mitochondria in living cells. Besides, MitoPDI-90 targets the mitochondria through electrical potential, also enabling live-cell MMP monitoring. MitoPDI-90 allows for super-resolution visualization and time-lapse imaging of mitochondria, and more importantly, indication of changes in MMP, providing insight into the functional activity of live-cell mitochondria.
Collapse
Affiliation(s)
- Lu Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rana Dalapati
- Department of Materials Science and Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Beibei Gao
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyu Huang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dan Zhao
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fu Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ling Zang
- Department of Materials Science and Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
30
|
Reigosa-Chamorro F, Cordeiro S, Pereira MT, Filipe B, Baptista PV, Fernandes AR, Vila JM. Effect of mono- and dinuclear thiosemicarbazone platinacycles in the proliferation of a colorectal carcinoma cell line. Dalton Trans 2024; 53:17803-17818. [PMID: 39233530 DOI: 10.1039/d4dt01490a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Herein, we describe the synthesis and characterization of a series of thiosemicarbazone platinacycles. Their activity towards HCT116 and A2780 cancer cell lines as well as normal fibroblasts was explored and conclusions about the influence of their structures were drawn based on the results. Ligands L1-3, tetranuclear compounds [Pt(L1-3)]4, [Pt(L1-3)(PPh3)], and [Pt(L1-L3)2{Ph2P(CH2)4PPh2}], and phosphine derivatives, were deemed unpromising owing to their lack of activity. However, mono-coordinated diphosphine complexes [Pt(L1-L3)(Ph2PCH2PPh2-P)] showed high selectivity and low IC50 values, and their antiproliferative activity was further studied. The three studied derivatives 3a, 3b and 3c showed a fast internalization of HCT116 colorectal cancer cells with similar IC50 values, which induced a depolarization of mitochondrial membrane potential, with the subsequent triggering of apoptosis and autophagy in the case of 3c. In the case of compounds 3a and 3b, cell death mechanisms (extrinsic and intrinsic apoptosis, respectively) were triggered via the induction of reactive oxygen species (ROS). The three compounds were not toxic to a chicken embryo in vivo (after 48 h), and, importantly, showed an anti-angiogenic potential after exposure to the IC50 of compounds 3a, 3b and 3c.
Collapse
Affiliation(s)
- Francisco Reigosa-Chamorro
- Departamento de Química Inorgánica, Universidade de Santiago de Compostela, Avenida das Ciencias s/n, 15782 Santiago de Compostela, Spain.
| | - Sandra Cordeiro
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal
| | - M Teresa Pereira
- Departamento de Química Inorgánica, Universidade de Santiago de Compostela, Avenida das Ciencias s/n, 15782 Santiago de Compostela, Spain.
| | - Beatriz Filipe
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal
| | - Pedro V Baptista
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal
| | - Alexandra R Fernandes
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal.
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Portugal
| | - José M Vila
- Departamento de Química Inorgánica, Universidade de Santiago de Compostela, Avenida das Ciencias s/n, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
31
|
Chen Z, Luo R, Xu T, Wang L, Deng S, Wu J, Wang H, Lin Y, Bu M. Design, synthesis and antitumor effects of lupeol quaternary phosphonium salt derivatives. Bioorg Med Chem 2024; 113:117934. [PMID: 39369566 DOI: 10.1016/j.bmc.2024.117934] [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: 08/08/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
Lupeol is a natural pentacyclic triterpenoid with a wide range of biological activities. To improve the water solubility and targeting of lupeol, in the following study, we synthesized 27 lupeol derivatives in the first series by introducing lipophilic cations with lupeol as the lead compound. Through the screening of different cancer cells, we found that some of the derivatives showed better activity than cisplatin against human non-small cell lung cancer A549 cells, among which compound 6c was found to have an IC50 value of 1.83 μM and a selectivity index of 21.02 (IC50MRC-5/IC50A549) against A549 cells. To further improve the antiproliferative activity of the compounds, we replaced the ester linkage of the linker with a carbamate linkage and synthesized a second series of five lupeol derivatives which were screened for activity, among which compound 14f was found to have an IC50 value of 1.36 μM and a selectivity index of 15.60 (IC50MRC-5/IC50A549) against A549 cells. We further evaluated the bioactivity of compounds 6c and 14f and found that both compounds induced apoptosis in A549 cells, promoted an increase in intracellular reactive oxygen species and decrease in mitochondrial membrane potential, and inhibited the cell cycle in the S phase. Of the compounds, compound 14f showed stronger bioactivity than compound 6c. We then selected compound 14f for molecular-level Western blot evaluation and in vivo evaluation in the zebrafish xenograft A549 tumor cell model. Compound 14f was found to significantly downregulate Bcl-2 protein expression and upregulate Bax, Cyt C, cleaved caspase-9, and cleaved caspase-3 protein expression, and 14f was found to be able to inhibit the proliferation of A549 cells in the zebrafish xenograft model. The above results suggest that compound 14f has great potential in the development of antitumor drugs targeting mitochondria.
Collapse
Affiliation(s)
- Zongxing Chen
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ran Luo
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Tianci Xu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jiale Wu
- College of Pharmacy, Hainan University, Haikou 570228, Hainan, PR China
| | - Haijun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| |
Collapse
|
32
|
Xu C, Huang X, Hu Q, Xue W, Zhou K, Li X, Nan Y, Ju D, Wang Z, Zhang X. Modulating autophagy to boost the antitumor efficacy of TROP2-directed antibody-drug conjugate in pancreatic cancer. Biomed Pharmacother 2024; 180:117550. [PMID: 39418963 DOI: 10.1016/j.biopha.2024.117550] [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: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Pancreatic cancer, characterized by a dismal prognosis and limited treatment options, persists as a formidable challenge in oncology. Trophoblast cell surface antigen 2 (TROP2)-directed antibody-drug conjugates have achieved great success in solid tumors such as breast cancer and uroepithelial carcinoma. However, their efficacy against pancreatic cancer was insufficient in clinical trials, necessitating an imperative exploration of underlying mechanisms and new therapeutic strategies. In this study, we indicated that αTROP2-MMAE, an antibody-drug conjugate targeting TROP2, induced apoptosis through the caspase-9/PARP pathway and exerted potent antitumor effects against TROP2-positive pancreatic cancer. Simultaneously, RNA sequencing suggested significant changes in autophagy after αTROP2-MMAE treatment. The formation of autophagosomes and activation of autophagic flux were markedly induced through mechanisms associated with suppressing the activation of the Akt/mTOR pathway. The addition of pharmacological inhibitors of autophagy enhanced the cytotoxicity and apoptosis caused by αTROP2-MMAE, revealing the cytoprotective role of autophagy in TROP2-positive pancreatic cancer. In the subcutaneous xenograft model using BxPC3 cells, the combined administration of αTROP2-MMAE and an autophagy inhibitor elevated the tumor inhibition rate of αTROP2-MMAE from 71.6 % to 99.0 %, resulting in the eradication of tumors in half of the mice. Collectively, our research demonstrated for the first time the cytoprotective role of autophagy in TROP2-targeted antibody-drug conjugate therapy for pancreatic cancer, providing new perspectives for mechanistic exploration and therapeutic strategies in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Caili Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiting Huang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qinchao Hu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenjing Xue
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Kaicheng Zhou
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xingxiu Li
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Ziyu Wang
- Department of Pharmacy, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
33
|
Ciszak L, Kosmaczewska A, Pawlak E, Frydecka I, Szteblich A, Wołowiec D. Association of Genetic Variants at the CDKN1B and CCND2 Loci Encoding p27 Kip1 and Cyclin D2 Cell Cycle Regulators with Susceptibility and Clinical Course of Chronic Lymphocytic Leukemia. Int J Mol Sci 2024; 25:11705. [PMID: 39519258 PMCID: PMC11546115 DOI: 10.3390/ijms252111705] [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: 10/02/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Beyond the essential role of p27Kip1 and cyclin D2 in cell cycle progression, they are also shown to confer an anti-apoptotic function in peripheral blood (PB) lymphocytes. Although the aberrant longevity and expression of p27Kip1 and cyclin D2 in leukemic cells is well documented, the exact mechanisms responsible for this phenomenon have yet to be elucidated. This study was undertaken to determine the associations between polymorphisms in the CDKN1B and CCND2 genes (encoding p27Kip1 and cyclin D2, respectively) and susceptibility to chronic lymphocytic leukemia (CLL), as well as their influence on the expression of both cell cycle regulators in PB leukemic B cells and non-malignant T cells from untreated CLL patients divided according to the genetic determinants studied. Three CDKN1B single-nucleotide polymorphisms (SNPs), rs36228499, rs34330, and rs2066827, and three CCND2 SNPs, rs3217933, rs3217901, and rs3217810, were genotyped using a real-time PCR system. The expression of p27Kip1 and cyclin D2 proteins in both leukemic B cells and non-malignant T cells was determined using flow cytometry. We found that the rs36228499A and rs34330T alleles in CDKN1B and the rs3217810T allele in the CCND2 gene were more frequent in patients and were associated with increased CLL risk. Moreover, we observed that patients possessing the CCND2rs3217901G allele had lower susceptibility to CLL (most pronounced in the AG genotype). We also noticed that the presence of the CDKN1Brs36228499CC, CDKN1Brs34330CC, CDKN1Brs2066827TT, and CCND2rs3217901AG genotypes shortened the time to CLL progression. Statistically significant functional relationships were limited to T cells and assigned to CDKN1B polymorphic variants; carriers of the polymorphisms rs34330CC and rs36228499CC (determining the aggressive course of CLL) expressed a decrease in p27Kip1 and cyclin D2 levels, respectively. We indicate for the first time that genetic variants at the CDKN1B and CCND2 loci may be considered as a potentially low-penetrating risk factor for CLL and determining the clinical outcome.
Collapse
Affiliation(s)
- Lidia Ciszak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (L.C.); (E.P.); (I.F.); (A.S.)
| | - Agata Kosmaczewska
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (L.C.); (E.P.); (I.F.); (A.S.)
| | - Edyta Pawlak
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (L.C.); (E.P.); (I.F.); (A.S.)
| | - Irena Frydecka
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (L.C.); (E.P.); (I.F.); (A.S.)
| | - Aleksandra Szteblich
- Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland; (L.C.); (E.P.); (I.F.); (A.S.)
| | - Dariusz Wołowiec
- Clinical Department of Hematology, Cell Therapies and Internal Diseases, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
34
|
Kazemeini S, Nadeem-Tariq A, Shih R, Rafanan J, Ghani N, Vida TA. From Plaques to Pathways in Alzheimer's Disease: The Mitochondrial-Neurovascular-Metabolic Hypothesis. Int J Mol Sci 2024; 25:11720. [PMID: 39519272 PMCID: PMC11546801 DOI: 10.3390/ijms252111720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) presents a public health challenge due to its progressive neurodegeneration, cognitive decline, and memory loss. The amyloid cascade hypothesis, which postulates that the accumulation of amyloid-beta (Aβ) peptides initiates a cascade leading to AD, has dominated research and therapeutic strategies. The failure of recent Aβ-targeted therapies to yield conclusive benefits necessitates further exploration of AD pathology. This review proposes the Mitochondrial-Neurovascular-Metabolic (MNM) hypothesis, which integrates mitochondrial dysfunction, impaired neurovascular regulation, and systemic metabolic disturbances as interrelated contributors to AD pathogenesis. Mitochondrial dysfunction, a hallmark of AD, leads to oxidative stress and bioenergetic failure. Concurrently, the breakdown of the blood-brain barrier (BBB) and impaired cerebral blood flow, which characterize neurovascular dysregulation, accelerate neurodegeneration. Metabolic disturbances such as glucose hypometabolism and insulin resistance further impair neuronal function and survival. This hypothesis highlights the interconnectedness of these pathways and suggests that therapeutic strategies targeting mitochondrial health, neurovascular integrity, and metabolic regulation may offer more effective interventions. The MNM hypothesis addresses these multifaceted aspects of AD, providing a comprehensive framework for understanding disease progression and developing novel therapeutic approaches. This approach paves the way for developing innovative therapeutic strategies that could significantly improve outcomes for millions affected worldwide.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (S.K.); (A.N.-T.); (R.S.); (J.R.); (N.G.)
| |
Collapse
|
35
|
Wu A, Zhao C. Astilbin Induces Apoptosis in Oral Squamous Cell Carcinoma through p53 Reactivation and Mdm-2 Inhibition. DOKL BIOCHEM BIOPHYS 2024; 518:429-441. [PMID: 39196525 DOI: 10.1134/s1607672924600374] [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: 04/23/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 08/29/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a frequently occurring malignancy in the head and neck region. The most commonly mutated gene in OSCC is the tumor suppressor gene p53 (TP53), linked to lower survival and treatment resistance in OSCC patients. Astilbin is a flavonoid amongst several herbal treatments with a variety of pharmacological actions mainly including antioxidant, anti-inflammatory, and anti-cancer characteristics. This study evaluated the effects of astilbin on proliferation of OSCC cell lines SCC90 and SCC4 (bearing a p53 mutation) in relevance to p53 and Mdm-2 pathways. Astilbin inhibited the proliferation of SCC4 and SCC90 cells in a dose- and time-dependent manner. The IC50 values for both the cell lines were about 75 μM for astilbin. A p53 activator (RITA) was used to determine the effects of astilbin on p53 activity, and the results demonstrated synergistic reduction in cell growth. However, when combined with pifithrin-α (a p53 inhibitor), astilbin demonstrated a strong inhibition of its response. Astilbin reduced the mitochondrial membrane potential in SCC4 cells, which is a sign of apoptotic activity. Astilbin decreased the amounts of Mdm-2 (negative regulator of p53) and increased the expression of the p53 gene and protein. In a p53-dependent manner, astilbin suppressed the ability of SCC4 cells to form colonies and heal wounds. This was followed by the induction of mitochondrial intrinsic apoptosis via the activation of caspases 9 and 3, cleavage of PARP, and the suppression of pro-apoptotic Bid. Astilbin-induced p53-mediated apoptosis in OSCC cells as herbal medicinal ingredients.
Collapse
Affiliation(s)
- Aimin Wu
- School of Medicine, JingChu University of Technology, 448000, JingMen, Hubei, China
| | - Chungang Zhao
- School of Medicine, JingChu University of Technology, 448000, JingMen, Hubei, China.
| |
Collapse
|
36
|
Yang RZ, Wang DD, Li SM, Liu PP, Kang JS. Development and Application of a Mitochondrial Genetically Encoded Voltage Indicator in Narcosis. Neurosci Bull 2024; 40:1529-1544. [PMID: 38829505 PMCID: PMC11422539 DOI: 10.1007/s12264-024-01235-w] [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: 09/27/2023] [Accepted: 01/11/2024] [Indexed: 06/05/2024] Open
Abstract
Mitochondrial membrane potential (MMP) plays a crucial role in the function of cells and organelles, involving various cellular physiological processes, including energy production, formation of reactive oxygen species (ROS), unfolded protein stress, and cell survival. Currently, there is a lack of genetically encoded fluorescence indicators (GEVIs) for MMP. In our screening of various GEVIs for their potential monitoring MMP, the Accelerated Sensor of Action Potentials (ASAP) demonstrated optimal performance in targeting mitochondria and sensitivity to depolarization in multiple cell types. However, mitochondrial ASAPs also displayed sensitivity to ROS in cardiomyocytes. Therefore, two ASAP mutants resistant to ROS were generated. A double mutant ASAP3-ST exhibited the highest voltage sensitivity but weaker fluorescence. Overall, four GEVIs capable of targeting mitochondria were obtained and named mitochondrial potential indicators 1-4 (MPI-1-4). In vivo, fiber photometry experiments utilizing MPI-2 revealed a mitochondrial depolarization during isoflurane-induced narcosis in the M2 cortex.
Collapse
Affiliation(s)
- Run-Zhou Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Dian-Dian Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Sen-Miao Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Pei-Pei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jian-Sheng Kang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
37
|
Gao Y, Chen Q, Yang S, Cao J, Li F, Li R, Wu Z, Wang Y, Yuan L. Indole alleviates nonalcoholic fatty liver disease in an ACE2-dependent manner. FASEB J 2024; 38:e70061. [PMID: 39305120 DOI: 10.1096/fj.202401172rr] [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: 05/25/2024] [Revised: 08/27/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
Indole is a microbial metabolite produced by the gut microbiota through the degradation of dietary tryptophan, known for its well-established anti-inflammatory and antioxidant properties. In this study, we collected fecal samples from mice fed a high-fat diet (HFD) and those on a standard diet (SD), then conducted 16S rRNA sequencing to analyze their gut microbiota. The analysis revealed distinct differences in the dominant bacterial species between the two groups, with a significant decrease in indole-producing probiotics in the HFD mice compared to the SD group. Then we administered oral indole treatment to male C57BL/6J mice with HFD-induced NAFLD and observed a significant improvement in hepatic steatosis and inflammation. Notably, indole alleviated the HFD-induced decline in serum Angiotensin-(1-7) [Ang-(1-7)] levels and Angiotensin-Converting Enzyme 2 (ACE2) expression. To further investigate the role of indole and ACE2 in NAFLD, we conducted experiments using ACE2 knockout (ACE2KO) mice that were also induced with HFD-induced NAFLD and treated with indole. Interestingly, the protective effects of indole were compromised in the absence of ACE2. In HepG2 cells, indole similarly stimulated ACE2 expression and, in an ACE2-dependent manner, reduced ROS generation, maintained mitochondrial membrane potential stability, and increased SIRT3 expression. In summary, our results highlight the formation of a biologically active gut-liver axis between the gut microbiota and the liver through the tryptophan metabolite indole, which mitigates NAFLD in an ACE2-dependent manner. Elevating dietary tryptophan and increasing indole levels may represent an effective approach for preventing and treating NAFLD.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songtao Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Cao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Zhang Y, Xie Y, Zhang X, Duan C, Ma J, Wang Y, Wu Y, Shan N, Cheng K, Zhuang R, Bian K. CD226 implicated in Akt-dependent apoptosis of CD4 + T cell contributes to asthmatic pathogenesis. Cell Death Dis 2024; 15:705. [PMID: 39349422 PMCID: PMC11442704 DOI: 10.1038/s41419-024-07080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/02/2024]
Abstract
Asthma is a chronic airway inflammatory disease in which CD4+ T cell dysregulation occurs. Here, we investigated the molecular role and clinical significance of CD226, a costimulatory molecule of T lymphocytes, in the development of allergic asthma. Our results revealed that the expression of CD226 was significantly increased in CD4+ effector T cells, especially in T helper (Th) 2 cells and Th17 cells in patients with asthma. Moreover, CD4+ T cell-specific Cd226-knockout mice were generated and together with littermates were challenged with ovalbumin (OVA) to establish a model of allergic asthma. We found that CD226 deficiency in CD4+ T cells mitigated lung inflammation, IgE production, and eosinophil infiltration and reduced airway remodeling in experimental allergic asthma. However, the impact of CD226 on asthma was independent of Treg cell modulation. Through RNA-seq data analysis, the apoptosis pathway was screened. Mechanistically, CD226 deletion promoted CD4+ T cell late apoptosis via the activation of Caspase-3 in an Akt-dependent manner. Furthermore, blocking CD226 signaling with a recombinant fusion protein attenuated asthma features in mice and achieved a good therapeutic effect. Overall, this study revealed a unique role of CD226 in CD4+ T cell regulation in asthma pathogenesis. Therefore, targeting CD226 may provide new insights into the clinical treatment of asthma.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yang Xie
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xuexin Zhang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jingchang Ma
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yilin Wu
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Niqi Shan
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ka Bian
- Department of Otolaryngology Head and Neck Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
39
|
Zhang Y, Bejaoui M, Linh TN, Arimura T, Isoda H. A novel amphiphilic squalene-based compound with open-chain polyethers reduces malignant melanoma metastasis in-vitro and in-vivo. Cell Commun Signal 2024; 22:437. [PMID: 39261954 PMCID: PMC11389383 DOI: 10.1186/s12964-024-01813-5] [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: 05/01/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Squalene (SQ) is a well-known antioxidant and anti-inflammatory agent that provides promising anti-aging and UV-protective roles on human skin. However, its strong hydrophobic nature, accompanied by issues such as poor solubility and limited tissue permeation, has created challenges for scientists to investigate its untapped potential in more complex conditions, including cancer progression. The present study assessed the potent anti-metastatic properties of a newly synthesized amphiphilic ethylene glycol SQ derivative (SQ-diEG) in melanoma, the most fatal skin cancer. In vitro and in vivo experiments have discovered that SQ-diEG may exert its potential on melanoma malignancy through the mitochondria-mediated caspase activation apoptotic signaling pathway. The potent anti-metastatic effect of SQ-diEG was observed in vitro using highly proliferative and aggressive melanoma cells. Administration of SQ-diEG (25 mg/kg) significantly decreased the tumor burden on the lung and inhibited the metastasis-associated proteins and gene markers in B16F10 lung colonization mice model. Furthermore, global gene profiling also revealed a promising role of SQ-diEG in tumor microenvironment. We anticipated that the amphiphilic nature of the SQ compound bearing ethylene glycol oligomers could potentially augment its ability to reach the pathology site, thus enhancing its therapeutic potential in melanoma.
Collapse
Affiliation(s)
- Yaman Zhang
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan
| | - Meriem Bejaoui
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Alliance for Research On the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan
| | - Tran Ngoc Linh
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Takashi Arimura
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Hiroko Isoda
- Tsukuba Life Science Innovation Program (T-LSI), University of Tsukuba, Tsukuba, Japan.
- Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.
- Alliance for Research On the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Japan.
- Institution of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
40
|
Badea MA, Balas M, Ionita D, Dinischiotu A. Carbon nanotubes conjugated with cisplatin activate different apoptosis signaling pathways in 2D and 3D-spheroid triple-negative breast cancer cell cultures: a comparative study. Arch Toxicol 2024; 98:2843-2866. [PMID: 38739308 PMCID: PMC11324667 DOI: 10.1007/s00204-024-03779-2] [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: 01/24/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024]
Abstract
The type of experimental model for the in vitro testing of drug formulations efficiency represents an important tool in cancer biology, with great attention being granted to three-dimensional (3D) cultures as these offer a closer approximation of the clinical sensitivity of drugs. In this study, the effects induced by carboxyl-functionalized single-walled carbon nanotubes complexed with cisplatin (SWCNT-COOH-CDDP) and free components (SWCNT-COOH and CDDP) were compared between conventional 2D- and 3D-spheroid cultures of human breast cancer cells. The 2D and 3D breast cancer cultures were exposed to various doses of SWCNT-COOH (0.25-2 μg/mL), CDDP (0.158-1.26 μg/mL) and the same doses of SWNCT-COOH-CDDP complex for 24 and 48 h. The anti-tumor activity, including modulation of cell viability, oxidative stress, proliferation, apoptosis, and invasion potential, was explored by spectrophotometric and fluorometric methods, immunoblotting, optical and fluorescence microscopy. The SWCNT-COOH-CDDP complex proved to have high anti-cancer efficiency on 2D and 3D cultures by inhibiting cell proliferation and activating cell death. A dose of 0.632 μg/mL complex triggered different pathways of apoptosis in 2D and 3D cultures, by intrinsic, extrinsic, and reticulum endoplasmic pathways. Overall, the 2D cultures showed higher susceptibility to the action of complex compared to 3D cultures and SWCNT-COOH-CDDP proved enhanced anti-tumoral activity compared to free CDDP.
Collapse
Affiliation(s)
- Madalina Andreea Badea
- Faculty of Biology, Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095, Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 90-92 Sos. Panduri, 050663, Bucharest, Romania
| | - Mihaela Balas
- Faculty of Biology, Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095, Bucharest, Romania.
| | - Daniela Ionita
- Faculty of Applied Chemistry and Materials Science, Department of General Chemistry, Politehnica University of Bucharest, 313 Splaiul Independentei, 060042, Bucharest, Romania
| | - Anca Dinischiotu
- Faculty of Biology, Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095, Bucharest, Romania
| |
Collapse
|
41
|
Markatos C, Biniari G, Chepurny OG, Karageorgos V, Tsakalakis N, Komontachakis G, Vlata Z, Venihaki M, Holz GG, Tselios T, Liapakis G. Cytotoxic Activity of Novel GnRH Analogs Conjugated with Mitoxantrone in Ovarian Cancer Cells. Molecules 2024; 29:4127. [PMID: 39274973 PMCID: PMC11397358 DOI: 10.3390/molecules29174127] [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: 04/30/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
The gonadotropin-releasing hormone (GnRH) receptor (GnRH-R) is highly expressed in ovarian cancer cells (OCC), and it is an important molecular target for cancer therapeutics. To develop a new class of drugs targeting OCC, we designed and synthesized Con-3 and Con-7 which are novel high-affinity GnRH-R agonists, covalently coupled through a disulfide bond to the DNA synthesis inhibitor mitoxantrone. We hypothesized that Con-3 and Con-7 binding to the GnRH-R of OCC would expose the conjugated mitoxantrone to the cellular thioredoxin, which reduces the disulfide bond of Con-3 and Con-7. The subsequent release of mitoxantrone leads to its intracellular accumulation, thus exerting its cytotoxic effects. To test this hypothesis, we determined the cytotoxic effects of Con-3 and Con-7 using the SKOV-3 human OCC. Treatment with Con-3 and Con-7, but not with their unconjugated GnRH counterparts, resulted in the accumulation of mitoxantrone within the SKOV-3 cells, increased their apoptosis, and reduced their proliferation, in a dose- and time-dependent manner, with half-maximal inhibitory concentrations of 0.6-0.9 µM. It is concluded that Con-3 and Con-7 act as cytotoxic "prodrugs" in which mitoxantrone is delivered in a GnRH-R-specific manner and constitute a new class of lead compounds for use as anticancer drugs targeting ovarian tumors.
Collapse
Affiliation(s)
- Christos Markatos
- Department of Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (C.M.); (V.K.); (N.T.); (G.K.)
| | - Georgia Biniari
- Department of Chemistry, University of Patras, 26504 Rion, Greece;
| | - Oleg G. Chepurny
- Department of Medicine, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA;
| | - Vlasios Karageorgos
- Department of Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (C.M.); (V.K.); (N.T.); (G.K.)
| | - Nikos Tsakalakis
- Department of Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (C.M.); (V.K.); (N.T.); (G.K.)
| | - Georgios Komontachakis
- Department of Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (C.M.); (V.K.); (N.T.); (G.K.)
| | - Zacharenia Vlata
- Flow Cytometry Facility, Institute of Molecular Biology and Biotechnology of the Foundation for Research and Technology Hellas (IMBB-FORTH), 70013 Heraklion, Greece;
| | - Maria Venihaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - George G. Holz
- Department of Medicine and Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY 13210, USA;
| | - Theodore Tselios
- Department of Chemistry, University of Patras, 26504 Rion, Greece;
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (C.M.); (V.K.); (N.T.); (G.K.)
| |
Collapse
|
42
|
Silva-Carvalho AÉ, Bispo ECI, da Silva IGM, Correa JR, Carvalho JL, Gelfuso GM, Saldanha-Araujo F. Characterization of ibrutinib's effects on the morphology, proliferation, phenotype, viability, and anti-inflammatory potential of adipose-derived mesenchymal stromal cells. Sci Rep 2024; 14:19906. [PMID: 39191849 DOI: 10.1038/s41598-024-71054-6] [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: 01/25/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
Ibrutinib (IB) is a tyrosine kinase inhibitor (TKI) that has immunomodulatory action and can be used as second-line therapy for steroid-refractory or steroid-resistant chronic Graft versus Host Disease (cGVHD). Mesenchymal stromal cells (MSCs) are distributed throughout the body and their infusion has also been explored as a second-line therapeutic alternative for the treatment of cGVHD. Considering the currently unknown effects of IB on endogenous MSCs, as well as the possible combined use of IB and MSCs for cGVHD, we investigated whether adipose tissue-derived MSCs present IB-targets, as well as the consequences of treating MSCs with this drug, regarding cell viability, proliferation, phenotype, and anti-inflammatory potential. Interestingly, we show for the first time that MSCs express several IB target genes. Also of note, the treatment of such cells with this TKI elevated the levels of CD90 and CD105 surface proteins, as well as VCAM-1. Furthermore, IB-treated MSCs presented increased mRNA expression of the anti-inflammatory genes PD-L1, TSG-6, and IL-10. However, continued exposure to IB, even at low doses, compromised the viability of MSCs. These data indicate that the use of IB can stimulate an anti-inflammatory profile in MSCs, but also that a continued exposure to IB can compromise MSC viability over time.
Collapse
Affiliation(s)
- Amandda Évelin Silva-Carvalho
- Laboratório de Hematologia E Células-Tronco, Departamento de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, DF, Brasil
- Laboratório de Farmacologia Molecular, Universidade de Brasília, Brasília, Brasil
| | - Elizabete Cristina Iseke Bispo
- Laboratório de Hematologia E Células-Tronco, Departamento de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, DF, Brasil
| | | | - José Raimundo Correa
- Laboratório de Microscopia E Microanálises, Universidade de Brasília, Brasília, Brasil
| | - Juliana Lott Carvalho
- Laboratório Multidisciplinar de Biociências, Universidade de Brasília, Brasília, Brasil
| | | | - Felipe Saldanha-Araujo
- Laboratório de Hematologia E Células-Tronco, Departamento de Ciências da Saúde, Universidade de Brasília, Campus Darcy Ribeiro, Brasília, DF, Brasil.
| |
Collapse
|
43
|
Muhmood A, Liu J, Liu D, Liu S, Azzam MM, Junaid MB, Hou L, Le G, Huang K. Mitigation of Deoxynivalenol (DON)- and Aflatoxin B1 (AFB1)-Induced Immune Dysfunction and Apoptosis in Mouse Spleen by Curcumin. Toxins (Basel) 2024; 16:356. [PMID: 39195766 PMCID: PMC11359138 DOI: 10.3390/toxins16080356] [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: 07/19/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
In the context of the potential immunomodulatory properties of curcumin in counteracting the detrimental effects of concurrent exposure to Deoxynivalenol (DON) and Aflatoxin B1 (AFB1), a comprehensive 28-days trial was conducted utilizing 60 randomly allocated mice divided into four groups. Administration of curcumin at a dosage of 5 mg/kg body weight in conjunction with DON at 0.1 mg/kg and AFB1 at 0.01 mg/kg body weight was undertaken to assess its efficacy. Results indicated that curcumin intervention demonstrated mitigation of splenic structural damage, augmentation of serum immunoglobulin A (IgA) and immunoglobulin G (IgG) levels, elevation in T lymphocyte subset levels, and enhancement in the mRNA expression levels of pro-inflammatory cytokines TNF-α, IFN-γ, IL-2, and IL-6. Furthermore, curcumin exhibited a suppressive effect on apoptosis in mice, as evidenced by decreased activity of caspase-3 and caspase-9, reduced expression levels of pro-apoptotic markers Bax and Cytochrome-c (Cyt-c) at both the protein and mRNA levels, and the maintenance of a balanced expression ratio of mitochondrial apoptotic regulators Bax and Bcl-2. Collectively, these findings offer novel insights into the therapeutic promise of curcumin in mitigating immunosuppression and apoptotic events triggered by mycotoxin co-exposure.
Collapse
Affiliation(s)
- Azhar Muhmood
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jianxin Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Dandan Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuiping Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mahmoud M. Azzam
- Animal Production Department, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Muhammad Bilawal Junaid
- Department of Plant Production, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Lili Hou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Guannan Le
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (A.M.); (J.L.); (D.L.); (S.L.); (L.H.); (G.L.)
- Institute of Animal Nutritional Health, Nanjing Agricultural University, Nanjing 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
44
|
Hong CE, Lyu SY. Modulation of Breast Cancer Cell Apoptosis and Macrophage Polarization by Mistletoe Lectin in 2D and 3D Models. Int J Mol Sci 2024; 25:8459. [PMID: 39126027 PMCID: PMC11313472 DOI: 10.3390/ijms25158459] [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: 06/27/2024] [Revised: 07/25/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Korean mistletoe (Viscum album L. var. coloratum) is renowned for its medicinal properties, including anti-cancer and immunoadjuvant effects. This study aimed to elucidate the mechanisms by which Korean mistletoe lectin (V. album L. var. coloratum agglutinin; VCA) modulates breast cancer cell apoptosis and macrophage polarization. The specific objectives were to (1) investigate the direct effects of VCA on MCF-7 breast cancer cells and THP-1-derived M1/M2 macrophages; (2) analyze the impact of VCA on the paracrine interactions between these cell types; and (3) compare the efficacy of VCA in 2D vs. 3D co-culture models to bridge the gap between in vitro and in vivo studies. We employed both 2D and 3D models, co-culturing human M1/M2 macrophages with human MCF-7 breast cancer cells in a Transwell system. Our research demonstrated that M1 and M2 macrophages significantly influenced the immune and apoptotic responses of breast cancer cells when exposed to VCA. M1 macrophages exhibited cytotoxic characteristics and enhanced VCA-induced apoptosis in both 2D and 3D co-culture models. Conversely, M2 macrophages initially displayed a protective effect by reducing apoptosis in breast cancer cells, but this protective effect was reversed upon exposure to VCA. Furthermore, our findings illustrate VCA's ability to modulate M1 and M2 polarization in breast cancer cells. Finally, the use of magnetic 3D cell cultures suggests their potential to yield results comparable to conventional 2D cultures, bridging the gap between in vitro and in vivo studies.
Collapse
Affiliation(s)
- Chang-Eui Hong
- College of Pharmacy, Sunchon National University, Suncheon 57922, Republic of Korea;
- Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Su-Yun Lyu
- College of Pharmacy, Sunchon National University, Suncheon 57922, Republic of Korea;
- Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Suncheon 57922, Republic of Korea
| |
Collapse
|
45
|
Yau JNN, Yempala T, Muthuramalingam RPK, Giustarini G, Teng G, Ang WH, Gibson D, Adriani G, Pastorin G. Fluorescence-Guided Spatial Drug Screening in 3D Colorectal Cancer Spheroids. Adv Healthc Mater 2024; 13:e2400203. [PMID: 38774999 DOI: 10.1002/adhm.202400203] [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: 01/18/2024] [Revised: 05/06/2024] [Indexed: 06/04/2024]
Abstract
The limited recapitulation of critical cancer features in 2D cultures causes poor translatability of preclinical results from in vitro assays to in vivo tumor models. This contributes to slow drug development with a low success rate. 3D cultures better recapitulate the tumor microenvironment, enabling more accurate predictions when screening drug candidates and improving the development of chemotherapeutics. Platinum (Pt) (IV) compounds are promising prodrugs designed to reduce the severe systemic toxicity of widely used Food and Drug Administration (FDA)-approved Pt(II) drugs such as cisplatin. Here, this work presents spatiotemporal evaluations in 3D colorectal cancer (CRC) spheroids of mitochondria-targeting Pt(IV) complexes. CRC spheroids provide a greater pathophysiological recapitulation of in vivo tumors than 2D cultures by a marked upregulation of the ABCG2 chemoresistance marker expression. Furthermore, new 3D-staining protocols are introduced to evaluate the real-time decrease in mitochondria membrane potential (ΔΨ) in CRC spheroids, and a Pt-sensing dye to quantify the Pt mitochondrial accumulation. Finally, this work demonstrates a correlation between in vitro results and the efficacy of the compounds in vivo. Overall, the CRC spheroids represent a fast and cost-effective model to assess the behavior of Pt compounds in vitro and predict their translational potential in CRC treatment.
Collapse
Affiliation(s)
- Jia Ning Nicolette Yau
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore
| | - Thirumal Yempala
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Ram Pravin Kumar Muthuramalingam
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Giulio Giustarini
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
| | - Germaine Teng
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
| | - Wee Han Ang
- Department of Chemistry, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
| | - Dan Gibson
- Institute for Drug Research, School of Pharmacy, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Giulia Adriani
- Singapore Immunology Network, Agency for Science, Technology, and Research, Singapore, 138648, Singapore
- Department of Biomedical Engineering, Faculty of Engineerin, National University of Singapore, Singapore, 117578, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117544, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
46
|
Chota A, Abrahamse H, George BP. Green synthesis and characterization of AgNPs, liposomal loaded AgNPs and ZnPcS 4 photosensitizer for enhanced photodynamic therapy effects in MCF-7 breast cancer cells. Photodiagnosis Photodyn Ther 2024; 48:104252. [PMID: 38901719 DOI: 10.1016/j.pdpdt.2024.104252] [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: 05/28/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Breast cancer remains a formidable challenge in oncology despite significant advancements in treatment modalities. Conventional therapies such as surgery, chemotherapy, radiation therapy, and hormonal therapy have been the mainstay in managing breast cancer for decades. However, a subset of patient's experiences treatment failure, leading to disease recurrence and progression. Therefore, this study investigates the therapeutic potential of green-synthesized silver nanoparticles (AgNPs) using an African medicinal plant (Dicoma anomala methanol root extract) as a reducing agent for combating breast cancer. AgNPs were synthesized using the bottom-up approach and later modified with liposomes (Lip) loaded with photosensitizer (PS) zinc phthalocyanine tetrasulfonate (Lip@ZnPcS4) using thin film hydration method. The successful formation and Lip modification of AgNPs, alongside ZnPcS4, were confirmed through various analytical techniques including UV-Vis spectroscopy, Fourier-transform infrared spectroscopy (FT-IR), high-resolution transmission electron microscopy (HR-TEM), scanning electron microscopy (SEM) and energy dispersive X-ray spectroscopy (EDS). Following a 24 h treatment period, MCF-7 cells were assessed for viability using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT viability assay), cell death analysis using mitochondrial membrane potential (MMP) (ΔΨm), Annexin V-fluorescein isothiocyanate (FITC)-propidium iodide (PI) kit, and caspase- 3, 8 and 9 activities. The experiments were repeated four times (n = 4), and the results were analyzed using SPSS statistical software version 27, with a confidence interval set at 0.95. The synthesized nanoparticles and nanocomplex, including AgNPs, AgNPs-Lip, Lip@ZnPcS4, and AgNPs-Lip@ZnPcS4, exhibited notable cytotoxicity and therapeutic efficacy against MCF-7 breast cancer cells. Notably, the induction of apoptosis, governed by the upregulation of apoptotic proteins i.e., caspase 8 and 9 activities. In addition, caspase 3 was not expressed by MCF-7 cells in both control and experimental groups. Given the challenging prognosis associated with breast cancer, the findings underscore the promise of liposomal nanoformulations in cancer photodynamic therapy (PDT), thus warranting further exploration in clinical settings.
Collapse
Affiliation(s)
- Alexander Chota
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Blassan P George
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa.
| |
Collapse
|
47
|
Dhanalakshmi B, Anil Kumar BM, Srinivasa Murthy V, Srinivasa SM, Vivek HK, Sennappan M, Rangappa S. Design, synthesis and docking studies of novel 4-aminophenol-1,2,4-oxadiazole hybrids as apoptosis inducers against triple negative breast cancer cells targeting MAP kinase. J Biomol Struct Dyn 2024; 42:5841-5857. [PMID: 37529915 DOI: 10.1080/07391102.2023.2239912] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/17/2023] [Indexed: 08/03/2023]
Abstract
In our study, a series of novel 4-aminophenol benzamide-1,2,4-oxadiazole hybrid analogues have been designed and synthesized by condensing 4-hydroxyphenyl arylamides (3a-c) and 5-chloromethyl-3-aryl-1,2,4-oxadiazoles (6a-d). The structure of the synthesised compounds was verified by various spectroscopic techniques (1H NMR, 13C NMR, IR and LC-MS). All the prepared compounds were subjected to in silico and in vitro antiproliferative study against TNBC cell lines MDA-MB-468 and MDA-MB-231. The investigations revealed that compound 7k significantly promoted apoptosis against MDA-MB-468 and MDA-MB-231 cells with IC50 values of 22.31 µM and 26.27 µM, respectively. Compound 7k interacted with crucial active sites of MAPK and exhibited the highest docking score of -7.06 kcal/mol. Docking was validated with molecular dynamic studies with simulation for 100 ns, depicting various stable interactions with MAPK. Consequently, 7k forms stable H-Bonds and π-π stacking with amino acid residues along with π-cation. Our investigations reveal that the in vitro antiproliferative study of 7k was in good correlation with the in silico studies. Hence, 7k serves as a potential novel lead for the inhibition of TNBCs by downregulating MAPK P38.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Boregowda Dhanalakshmi
- Department of Chemistry, School of Engineering, Dayananda Sagar University, Bengaluru, Karnataka, India
- Department of Chemistry, Rajeev Institute of Technology, Visvesvaraya Technological University, Hassan, Karnataka, India
| | - Belagal Motatis Anil Kumar
- Department of Molecular Biology, Adichunchanagiri School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT, B.G Nagara, Karnataka, India
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, B.G Nagara, Karnataka, India
| | | | - Sudhanva Muddenahalli Srinivasa
- Department of Molecular Biology, Adichunchanagiri School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT, B.G Nagara, Karnataka, India
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, B.G Nagara, Karnataka, India
| | - Hamse Kameshwar Vivek
- Department of Biochemistry, Adichunchanagiri School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT, B.G Nagara, Karnataka, India
- Department of Biochemistry, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, B.G Nagara, Karnataka, India
| | - Madhappan Sennappan
- Department of Chemistry, Dayananda Sagar College of Engineering, Bangalore, Karnataka, India
| | - Shobith Rangappa
- Department of Molecular Biology, Adichunchanagiri School of Natural Sciences, ACU-CRI, Adichunchanagiri University, BGSIT, B.G Nagara, Karnataka, India
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, B.G Nagara, Karnataka, India
| |
Collapse
|
48
|
Li Z, Yu H, Hussain SA, Yang R. Anticancer activity of Araguspongine C via inducing apoptosis, and inhibition of oxidative stress, inflammation, and EGFR-TK in human lung cancer cells: An in vitro and in vivo study. J Biochem Mol Toxicol 2024; 38:e23763. [PMID: 38984790 DOI: 10.1002/jbt.23763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
The advanced non-small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutations has put a selective pressure on the discovery and development of newer EGFR inhibitors. Therefore, the present study intends to explore the pharmacological effect of Araguspongine C (Aragus-C) as anticancer agent against lung cancer. The effect of Aragus-C was evaluated on the viability of the A549 and H1975 cells. Further biochemical assays were performed to elaborate the effect of Aragus-C, on the apoptosis, cell-cycle analysis, and mitochondrial membrane potential in A549 cells. Western blot analysis was also conducted to determine the expression of EGFR in A549 cells. Tumor xenograft mice model from A549 cells was established to further elaborate the pharmacological activity of Aragus-C. Results suggest that Aragus C showed significant inhibitory activity against A549 cells as compared to H1975 cells. It has been found that Aragus-C causes the induction of apoptosis and promotes cell-cycle arrest at the G2/M phase of A549 cells. It also showed a reduction in the overexpression of EGFR in A549 cells. In tumor xenograft mice model, it showed a significant reduction of tumor volume in a dose-dependent manner, with maximum inhibitory activity was reported by the 8 mg/kg treated group. It also showed significant anti-inflammatory and antioxidant activity by reducing the level of TNF-α, IL-1β, IL-6, and MDA, with a simultaneous increase of superoxide dismutase and glutathione peroxidase. We have demonstrated the potent anti-lung cancer activity of Aragus-C, and it may be considered as a potential therapeutic choice for NSCLC treatment.
Collapse
Affiliation(s)
- Zhe Li
- Department of Oncology and Hematology, Yan'an People's Hospital, Yan'an, China
| | - Hongjiang Yu
- Department of Medical Oncology, Tongliao City Hospital, Tongliao, China
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Rui Yang
- Department of Medical Oncology, Yan'an People's Hospital, Yan'an, China
| |
Collapse
|
49
|
Jiang L, Liu Y, Tumbath S, Boudreau MW, Chatkewitz LE, Wang J, Su X, Zahid KR, Li K, Chen Y, Yang K, Hergenrother PJ, Huang X. Isopentyl-Deoxynboquinone Induces Mitochondrial Dysfunction and G2/M Phase Cell Cycle Arrest to Selectively Kill NQO1-Positive Pancreatic Cancer Cells. Antioxid Redox Signal 2024; 41:74-92. [PMID: 37950707 PMCID: PMC11321107 DOI: 10.1089/ars.2022.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 11/13/2023]
Abstract
Aims: Pancreatic cancer is among the top five leading causes of cancer-related deaths worldwide, with poor overall survival rates. Current therapies for pancreatic cancer lack tumor specificity, resulting in harmful effects on normal tissues. Therefore, developing tumor-specific agents for the treatment of pancreatic cancer is critical. NAD(P)H:quinone oxidoreductase 1 (NQO1), highly expressed in pancreatic cancers but not in associated normal tissues, makes NQO1 bioactivatable drugs a potential therapy for selectively killing NQO1-positive cancer cells. Our previous studies have revealed that the novel NQO1 bioactivatable drug deoxynyboquinone (DNQ) is 10-fold more potent than the prototypic NQO1 bioactivatable drug β-lapachone in killing of NQO1-positive cancer cells. However, DNQ treatment results in high-grade methemoglobinemia, a significant side effect that limits clinical development. Results: Here, we report for the first time on a DNQ derivative, isopentyl-deoxynboquinone (IP-DNQ), which selectively kills pancreatic ductal adenocarcinoma (PDAC) cells in an NQO1-dependent manner with equal potency to the parent DNQ. IP-DNQ evokes massive reactive oxygen species (ROS) production and oxidative DNA lesions that result in poly(ADP-ribose)polymerase-1 (PARP1) hyperactivation, mitochondrial catastrophe, and G2/M phase cell cycle arrest, leading to apoptotic and necrotic programmed cell death. Importantly, IP-DNQ treatment causes only mild methemoglobinemia in vivo, with a threefold improvement in the maximum tolerated dose (MTD) compared with DNQ, while it significantly suppresses tumor growth and extends the life span of mice in subcutaneous and orthotopic pancreatic cancer xenograft models. Innovation and Conclusion: Our study demonstrates that IP-DNQ is a promising therapy for NQO1-positive pancreatic cancers and may enhance the efficacy of other anticancer drugs. IP-DNQ represents a novel approach to treating pancreatic cancer with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Lingxiang Jiang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yingchun Liu
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P.R. China
| | - Soumya Tumbath
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matthew W. Boudreau
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Lindsay E. Chatkewitz
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jiangwei Wang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xiaolin Su
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kashif Rafiq Zahid
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Katherine Li
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kai Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- The Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul J. Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
50
|
Zhao H, Song J, Wang T, Fan X. Selenium nanoparticles decorated with polysaccharides from Sargassum fusiforme protects against 6-OHDA-induced neurotoxicity in PC12 cells and rat model of Parkinson's disease. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 59:102755. [PMID: 38762132 DOI: 10.1016/j.nano.2024.102755] [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: 11/18/2023] [Revised: 04/15/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder and identifying disease-causing pathways and drugs that target them has remained challenging. Herein, selenium nanoparticles decorated with polysaccharides from Sargassum fusiforme (SFPS-SeNPs) were investigated on 6-OHDA-induced neurotoxicity in PC12 cells and rats. 6-OHDA can significantly increase neurotoxicity, oxidative stress and decrease the activity of superoxide dismutase (SOD) and glutathione peroxidase (GPx) both in vitro and vivo. In vitro, treatment with SFPS-SeNPs can significantly decrease 6-OHDA cytotoxicity, reactive oxygen species (ROS) production or malondialdehyde (MDA) levels, and cell apoptosis, significantly increased the activity of SOD and GPx. In vivo, 6-OHDA exposure could also decrease the expression of Nrf2 and OH-1, while treatment with SFPS-SeNPs (1 mg Se/kg) increased. SFPS-SeNPs can protect neurons from 6-OHDA-induced neurotoxicity by regulating apoptosis and Nrf2/ARE pathway. The present study demonstrated that SFPS-SeNPs is a good candidate for developing a new drug against neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Hongying Zhao
- South China University of Technology, College of Food Science & Engineering, 381 Wushan Road, Guangzhou 510640, China.
| | - Jiaxin Song
- South China University of Technology, College of Food Science & Engineering, 381 Wushan Road, Guangzhou 510640, China.
| | - Tian Wang
- South China University of Technology, College of Food Science & Engineering, 381 Wushan Road, Guangzhou 510640, China.
| | - Xiaodan Fan
- South China University of Technology, College of Food Science & Engineering, 381 Wushan Road, Guangzhou 510640, China.
| |
Collapse
|