1
|
Mohamed HRH, Hakeem GM, Latif YA, Elnawasani SH, Nagy M, Mohamed BA, Essam R, Safwat G. Yttrium oxide nanoparticles induce selective cytotoxicity, genomic instability and ROS mitochondrial P53 mediated apoptosis in human pancreatic cancer cells. Sci Rep 2025; 15:20144. [PMID: 40542009 DOI: 10.1038/s41598-025-05088-9] [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: 01/09/2025] [Accepted: 05/30/2025] [Indexed: 06/22/2025] Open
Abstract
Pancreatic cancer is a hard-to-treat tumor with a poor prognosis. While traditional pancreatic cancer therapies can be effective, issues like cytotoxicity, low selectivity, and drug resistance still pose major challenges. Nanotechnology has shown promise in improving cancer diagnosis and treatment. Yttrium oxide nanoparticles (Y2O3-NPs), for example, have demonstrated potent selective cytotoxicity against triple negative breast cancer cells; but their effects on pancreatic cancer cells have not been explored. This study aimed to explore the impact of Y2O3-NPs on cell proliferation, DNA integrity, and oxidative stress in pancreatic cancer (PANC-1) and human skin fibroblast (HSF) cells. The cytotoxicity of Y2O3-NPs after 72 h were estimated using Sulforhodamine (SRB) cytotoxicity assay, while alkaline Comet assay was done to study genomic DNA integrity. Generation level of reactive oxygen species (ROS) and integrity of mitochondrial membrane potential were also analyzed. Apoptosis induction was investigated using Flow Cytometry and expression level of apoptotic (p53), anti-apoptotic (Bcl2) and mitochondrial (ND3) genes was measured using quantitative RTPCR. Our findings exhibited that Y2O3-NPs had strong selective cytotoxicity against PANC-1 cells with an IC50 value of 31.06 µg/ml, while having minimal effect on normal HSF cells (IC50 = 319.21 µg/ml). Treatment of PANC-1 cells with Y2O3-NPs at the IC50 concentration for 72 h significantly increased intracellular ROS levels and DNA damage, along with a notable reduction in mitochondrial membrane potential. Additionally, a significant rise in necrotic, early, and late apoptotic cells was observed, accompanied by downregulation of the anti-apoptotic Bcl2 gene and upregulation of the apoptotic p53 and mitochondrial ND3 genes. These findings highlight the selective toxicity of Y2O3-NPs towards cancerous PANC-1 cells, with minimal impact on normal cells. Y2O3-NPs appear to induce apoptosis in cancer cells by increasing ROS generation, damaging DNA, disrupting mitochondrial function, and triggering cell death. This study suggests that Y2O3-NPs may be a promising candidate for pancreatic cancer treatment. Further research is needed to fully explore their therapeutic potential.
Collapse
Affiliation(s)
- Hanan R H Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - George M Hakeem
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Yasmin Abdel Latif
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Shahd H Elnawasani
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Maria Nagy
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Basma A Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Rawan Essam
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| | - Gehan Safwat
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), 6th of October City, Egypt
| |
Collapse
|
2
|
Joung H, Seo S, Liu H. MG132 induces cell type‑specific anticancer effects in uterine leiomyosarcoma cell lines. Mol Med Rep 2025; 31:159. [PMID: 40211694 PMCID: PMC12015380 DOI: 10.3892/mmr.2025.13524] [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: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/25/2025] Open
Abstract
Uterine leiomyosarcoma (Ut‑LMS) is a rare and aggressive malignant tumor with limited therapeutic options. Therefore, exploration of novel treatment strategies is necessary. MG132 is a potent proteasome inhibitor that has shown promising potential in cancer therapy by inducing apoptosis through disruption of protein homeostasis. Despite its promising applications in various cancers, its effects on Ut‑LMS remains largely unexplored. Therefore, the present study investigated the effects of MG132 on Ut‑LMS cell lines (SK‑LMS‑1, SK‑UT‑1 and SK‑UT‑1B) in terms of cytotoxicity, apoptosis induction, cell cycle progression, autophagy and reactive oxygen species (ROS) production. Treatment with MG132 (0‑2 µM for 24 h) induced a dose‑dependent reduction in cell viability across all three cell lines, and the lactate dehydrogenase release assays confirmed membrane damage. Moreover, apoptosis induction was assessed using annexin V and 7‑AAD staining, which revealed dose‑dependent apoptosis in all three cell lines. Western blot analysis revealed increased cleaved poly‑adenosine diphosphate ribose polymerase and caspase‑3 levels, thereby indicating activation of the apoptotic pathway in response to MG132 treatment. MG132 also induced G2/M phase arrest in SK‑LMS‑1 and SK‑UT‑1 cells and altered the expression of cell cycle regulatory proteins, such as p21, p27 and p53. Furthermore, MG132 promoted autophagy in all three cell lines by increasing light chain 3 II levels. ROS levels remained unchanged in SK‑LMS‑1 cells but increased in SK‑UT‑1B and SK‑UT‑1 cells. Furthermore, the ROS scavenger N‑acetylcysteine effectively reduced MG132‑induced apoptosis in SK‑UT‑1 cells. These findings highlight the cytotoxicity of MG132 in Ut‑LMS cells, emphasize its potential as a therapeutic agent for Ut‑LMS, provide insights into its mechanisms of action, and suggest possible strategies for improving treatment efficacy.
Collapse
Affiliation(s)
- Hosouk Joung
- Research Institute of Medical Sciences, Chonnam National University Medical School, Hwasun-eup, Jeollanam-do 58128, Republic of Korea
| | - Suho Seo
- Department of Food and Drug, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Hyunju Liu
- Department of Obstetrics and Gynecology, Chosun University College of Medicine, Gwangju 61452, Republic of Korea
- Department of Obstetrics and Gynecology, Chosun University Hospital, Gwangju 61453, Republic of Korea
| |
Collapse
|
3
|
Elyasi L, Rosenholm JM, Jahanshahi M, Jesmi F. The Effect of Picein on Inhibitory Avoidance Memory and Activity of Antioxidant Enzymes in Hippocampus of Male Rats with Scopolamine-Induced Injury. Mol Neurobiol 2025; 62:7835-7845. [PMID: 39946000 DOI: 10.1007/s12035-025-04740-9] [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: 09/07/2024] [Accepted: 02/02/2025] [Indexed: 05/15/2025]
Abstract
Alzheimer disease (AD) is a common neurologic disorder, impairing memory and spatial perception. Consistent with the extensive search for its treatment, we investigated the effect of Picein on inhibitory avoidance memory, lipid peroxidation, and the activity of hippocampal antioxidant enzymes in rats. Forty adult male Wistar rats were randomized into control group (no intervention), model group (intraperitoneal injection of 3-mg/kg scopolamine), and three interventional groups (1.5-, 2.5-, and 5-mg/kg intraventricular Picein, once a day for 7 days, 24 h after scopolamine injection). After behavioral test, the rats' hippocampus was isolated for measuring oxidative stress markers, including enzymes superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GPX), catalase (CAT), and total antioxidant capacity (TAC). One-way ANOVA was used for comparing numeric variables among the groups using SPSS v.21. The results showed scopolamine decreased SOD, GPX, and CAT enzymes, and TAC level, and increased MDA level, compared with the control group (P < 0.001) that confirmed the scopolamine-induced AD model. The two doses of 2.5- and 5-mg/kg Picein increased latency for entering the dark room, compared to the scopolamine group (P < 0.05), making them similar to the control group. The number of entries into the dark room in the 2.5-mg/kg Picein reduced and approached the control group (P < 0.05). The 2.5-mg/kg Picein decreased MDA and increased SOD, GPX, and TAC, more than 5 mg/kg Picein, both different than scopolamine; only 2.5-mg/kg Picein had different CAT, compared to scopolamine group (P < 0.05). In conclusion, by lowering oxidative stress in the hippocampus, Picein was able to prevent the scopolamine-induced impaired learning and avoidance memory in rats.
Collapse
Affiliation(s)
- Leila Elyasi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Km 4 Gorgan-Sari Road (Shastcola), Gorgan, Iran.
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20500, Turku, Finland
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Km 4 Gorgan-Sari Road (Shastcola), Gorgan, Iran
| | - Fatemeh Jesmi
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Voros C, Stavros S, Sapantzoglou I, Mavrogianni D, Daskalaki MA, Theodora M, Antsaklis P, Drakakis P, Loutradis D, Daskalakis G. The Role of Placental Mitochondrial Dysfunction in Adverse Perinatal Outcomes: A Systematic Review. J Clin Med 2025; 14:3838. [PMID: 40507600 PMCID: PMC12155783 DOI: 10.3390/jcm14113838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/26/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
Background: Mitochondria are essential for placental function as they regulate energy metabolism, oxidative balance, and apoptotic signaling. Increasing evidence suggests that placental mitochondrial dysfunction may play a role in the development of many poor perinatal outcomes, including preeclampsia, intrauterine growth restriction (IUGR), premature birth, and stillbirth. Nonetheless, no systematic review has thoroughly investigated this connection across human research. This study aims to consolidate evidence from human research concerning the link between placental mitochondrial dysfunction and negative birth outcomes. Methods: A systematic search of PubMed, Scopus, and Web of Science identified human research examining placental mitochondrial features (e.g., mtDNA copy number, ATP production, oxidative stress indicators) in connection with adverse pregnancy outcomes. Methodological variety resulted in narrative data extraction and synthesis. Results: Twenty-nine studies met the inclusion criteria. Mitochondrial dysfunction was consistently associated with PE, IUGR, FGR, and PTB. The most often observed outcomes included diminished mtDNA copy number, decreased ATP production, elevated reactive oxygen species (ROS), and disrupted mitochondrial dynamics, characterized by increased DRP1 and decreased MFN2. Early-onset preeclampsia and symmetric fetal growth restriction exhibited particularly severe mitochondrial abnormalities, indicating a primary placental origin of the condition. Conclusions: A significant factor contributing to adverse pregnancy outcomes is the dysfunction of placental mitochondria. The analogous molecular signatures across many disorders suggest promising avenues for developing targeted therapies aimed at improving maternal-fetal health and predictive biomarkers.
Collapse
Affiliation(s)
- Charalampos Voros
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Sofoklis Stavros
- 3rd Department of Obstetrics and Gynecology, Attikon Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462 Chaidari, Greece;
| | - Ioakeim Sapantzoglou
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Despoina Mavrogianni
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Maria Anastasia Daskalaki
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Marianna Theodora
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Panagiotis Antsaklis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Peter Drakakis
- 3rd Department of Obstetrics and Gynecology, Attikon Hospital, National and Kapodistrian University of Athens, Rimini 1, 12462 Chaidari, Greece;
| | - Dimitrios Loutradis
- Fertility Institute-Assisted Reproduction Unit, Paster 15, 11528 Athens, Greece;
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Georgios Daskalakis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 Vasilissis Sofias Avenue, 11528 Athens, Greece; (I.S.); (D.M.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| |
Collapse
|
5
|
Thakkar AB, Subramanian RB, Thakkar VR, Thakkar SS, Prajapati J, Goswami D, Thakor P. Biochanin A, an isoflavone isolated from Dalbergia sissoo Roxb. ex DC., leaves promote ROS-mediated and caspase-dependent apoptosis in lung adenocarcinoma cells. J Biomol Struct Dyn 2025:1-25. [PMID: 40432355 DOI: 10.1080/07391102.2025.2507820] [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: 06/22/2023] [Accepted: 04/27/2024] [Indexed: 05/29/2025]
Abstract
The objective of this study was to isolate and characterize a cytotoxic compound from the hydromethanolic extract of Dalbergia sissoo Roxb. ex DC. leaves using the cold percolation technique. Thin-layer chromatography was employed to isolate the cytotoxic component from the crude plant extract, and its cytotoxicity against lung adenocarcinoma (A549) cells was evaluated using the MTT assay. The structure of the isolated cytotoxic compound was determined through FTIR, NMR, UV analysis, and LC-MS/MS methods. Through comprehensive characterization, a cytotoxic compound called Biochanin A (BA) was identified, exhibiting significant anticancer activity with an IC50 value of 21.92 ± 2.19 μM against A549 cells, while demonstrating lower cytotoxicity towards normal lung cells (WI-38) with an IC50 value of 285.12 ± 2.19 μM. Notably, BA induced morphological changes in A549 cells, leading to apoptotic alterations and the generation of reactive oxygen species (ROS), as confirmed by multiple techniques (AO/EB, DAPI, Giemsa). In silico molecular docking, ADMET, MMGBSA, and molecular dynamics simulation investigations support the RT-PCR and cell biology findings. As a result, BA's molecular mechanism of action involves ROS-induced apoptosis mediated by caspases 9 and 3.
Collapse
Affiliation(s)
- Anjali B Thakkar
- P. G. Department of Biosciences, Sardar Patel Maidan, Satellite Campus, Sardar Patel University, Anand, Gujarat, India
- Department of Applied and Interdisciplinary Sciences, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Ramalingam B Subramanian
- P. G. Department of Biosciences, Sardar Patel Maidan, Satellite Campus, Sardar Patel University, Anand, Gujarat, India
| | - Vasudev R Thakkar
- P. G. Department of Biosciences, Sardar Patel Maidan, Satellite Campus, Sardar Patel University, Anand, Gujarat, India
| | - Sampark S Thakkar
- Akashganga, Shree Kamdhenu Electronics Pvt. Ltd, Vallabh Vidyanagar, Gujarat, India
| | - Jignesh Prajapati
- Department of Biochemistry & Forensic Sciences, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Dweipayan Goswami
- Department of Microbiology & Biotechnology, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Parth Thakor
- Bapubhai Desaibhai Patel Institute of Paramedical Sciences, Charotar University of Science and Technology, Changa, Gujarat, India
| |
Collapse
|
6
|
Farias HR, Costa-Beber LC, Costa Rodrigues Guma FT, de Oliveira J. Hypercholesterolemia, oxidative stress, and low-grade inflammation: a potentially dangerous scenario to blood-brain barrier. Metab Brain Dis 2025; 40:205. [PMID: 40380979 DOI: 10.1007/s11011-025-01620-y] [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: 01/31/2024] [Accepted: 04/23/2025] [Indexed: 05/19/2025]
Abstract
For more than a century, hypercholesterolemia has been linked to atherosclerotic cardiovascular disease. Notably, this metabolic condition has also been pointed out as a risk factor for neurodegenerative diseases, such as Alzheimer's disease (AD). Oxidative stress seems to be the connective factor between hypercholesterolemia and cardio and neurological disorders. By disturbing redox homeostasis, hypercholesterolemia impairs nitric oxide (NO) availability, an essential vasoprotective element, and jeopardizes endothelial function and selective permeability. The central nervous system (CNS) is partially protected from peripheral insults due to an arrangement between endothelial cells, astrocytes, microglia, and pericytes that form the blood-brain barrier (BBB). The endothelial dysfunction related to hypercholesterolemia increases the risk of developing cardiovascular diseases and also initiates BBB breakdown, which is a cause of brain damage characterized by neuroinflammation, oxidative stress, mitochondrial dysfunction, and, ultimately, neuronal and synaptic impairment. In this regard, we reviewed the mechanisms by which hypercholesterolemia-induced oxidative stress affects peripheral vessels, BBB, and leads to memory deficits. Finally, we suggest oxidative stress as the missing link between hypercholesterolemia and dementia.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Lílian Corrêa Costa-Beber
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
7
|
Munusamy MA, Bharathi M, Alarfaj AA, Hussein-Al-Ali SH, Sarathbabu S. Pluronic F-127 coated zinc oxide nanoparticles from Gynura pseudochina: a comprehensive assessment of antibacterial, and anticancer activities. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:872-891. [PMID: 39673551 DOI: 10.1080/09205063.2024.2434307] [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: 09/03/2024] [Accepted: 10/30/2024] [Indexed: 12/16/2024]
Abstract
The study synthesized Pluronic F-127 coated zinc oxide nanoparticles (PF-127ZnO NPs) using Gynura pseudochina leaf extract and evaluated their antibacterial, antioxidant, and anticancer properties using various characterization methods such as UV-Vis, Fourier Transform Infra-Red, Photoluminescence spectroscopy, FESEM, EDAX, TEM, SAED, XRD, and DLS. The disc diffusion technique was used to evaluate the antibacterial properties of synthesized Pluronic F-127 coated zinc oxide nanoparticles against various pathogens and significant anticancer activity was noticed. The study examined the cytotoxicity of PF127ZnO nanoparticles to Hep3B cells in vitro. The cytotoxicity was IC50 at 10 µM concentration. The study examined the biocompatibility of PF-127ZnO NPs, revealing membrane blebbing, chromatin condensation, and stimulation of the proapoptotic protein caspase cascade family. The study reveals that PF-127ZnO NPs, synthesized from G. pseudochina leaf extract, exhibit antibacterial and antihepatic properties, offering potential biomedical applications due to their high stability and biocompatibility.
Collapse
Affiliation(s)
- Murugan Alwarkurichi Munusamy
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | | | - Subbarayan Sarathbabu
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Namakkal, Tamil Nadu, India
| |
Collapse
|
8
|
Hitti G, Kavanaugh A, Zukotynski B, Billi F. Management of FSHD symptoms: current assistive technologies and pharmacological approaches. Disabil Rehabil Assist Technol 2025; 20:733-741. [PMID: 39565698 DOI: 10.1080/17483107.2024.2431058] [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: 03/06/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Facioscapulohumeral Muscular Dystrophy (FSHD) is a genetically linked disorder characterized by the progressive deterioration of muscles controlling facial and scapular movement. The severity and distribution of affected muscle groups vary significantly across patient demographics, necessitating diverse assistive approaches. OBJECTIVE This review aims to evaluate the effectiveness of assistive devices and therapeutic options, including medications and rehabilitative therapies, tailored to specific manifestations of FSHD. METHODS An analysis of existing literature and clinical trials was conducted to explore current assistive technologies and pharmacological treatments, focusing on their application to FSHD patients with varying symptom severity. RESULTS Although several pharmacological treatments, such as steroids, supplements, protein inhibitors, and knockout strategies, are under investigation, none have yet fully counteracted the disease process driven by toxic DUX4 production. Consequently, the broad assortment of assistive devices currently on the market remain critical for improving quality of life. CONCLUSION Despite advances in pharmacological research, the variability in FSHD manifestations necessitates a personalized approach combining assistive technologies and tailored therapeutic interventions. Future research should continue exploring integrative strategies to address the unique needs of FSHD patients.
Collapse
Affiliation(s)
- George Hitti
- UCLA Orthopedic Surgery Research Center, Los Angeles, USA
| | | | | | - Fabrizio Billi
- UCLA Orthopedic Surgery Research Center, Los Angeles, USA
| |
Collapse
|
9
|
Sammari H, Abidi A, Jedidi S, Dhawefi N, Sebai H. Laxative Effect of Crataegus azarolus Leaves Decoction Extract Against Loperamide-Induced Constipation and Oxidative Stress in Rats. J Med Food 2025; 28:465-477. [PMID: 40107769 DOI: 10.1089/jmf.2023.0255] [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: 03/22/2025] Open
Abstract
Constipation represents a common gastrointestinal disorder that has various adverse effects on the gastrointestinal tract. As a result, various civilizations have developed phytomedicines in order to treat and relieve its symptoms. In the current study, we evaluated the effect of Crataegus azarolus L. leaves decoction extract (CALDE) against loperamide (LOP)-induced constipation in rats. For 3 days, treated rats were administered LOP (3 mg/kg, b.w., p.o.) and CALDE (50, 100, and 200 mg/kg, b.w., p.o.) or yohimbine (2 mg/kg, b.w., p.o.). The gastric emptying test or intestinal transit time was calculated. The oxidative status was studied and evaluated using biochemical colorimetric methods. Results showed that CALDE administration improves gastric emptying and accelerates gastrointestinal transit. Pretreatment with LOP altered the defecation parameters and generated an oxidative status in healthy rats. In contrast, CALDE coadministration protected against the deregulation of intestinal motor function and frequency of defecation and significantly re-established oxidative marker levels. CALDE treatment demonstrated significant protection against experimental oxidative stress and constipation induced by LOP. Therefore, it can be considered a pharmacological drug to treat these gastrointestinal troubles.
Collapse
Affiliation(s)
- Houcem Sammari
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Anouar Abidi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
- National Institute of Technologies and Sciences of Kef (INTeK), University of Jendouba, Kef, Tunisia
| | - Nourhène Dhawefi
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization, Higher Institute of Biotechnology of Béja, University of Jendouba, Béja, Tunisia
| |
Collapse
|
10
|
Miceli V, Lo Gerfo E, Russelli G, Bulati M, Iannolo G, Tinnirello R, Cimino M, Saso L, Avorio F, Lo Re V. Circulating Biomarkers to Predict Post-Operative Cognitive Decline in Patients Undergoing Coronary Artery Bypass Grafting. Cell Mol Neurobiol 2025; 45:37. [PMID: 40254644 PMCID: PMC12009791 DOI: 10.1007/s10571-025-01553-1] [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: 02/17/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
Post-operative cognitive decline (POCD) is characterized by impairments in cognitive functions. Coronary artery bypass grafting (CABG) is associated with a high risk of POCD due to its impact on neuroinflammation and oxidative stress. In this study, we investigated the dynamics of neurotrophic, inflammatory, and oxidative stress markers in a cohort of post-CABG patients to identify potential biomarkers for POCD. Blood samples were collected at baseline (immediately post-surgery) and at 3-month follow-up. Expression levels of NRF2 and other regulators of oxidative stress (GST, GSS, HMOX1, CAT, HSP27, and LOX-1), inflammatory mediators (IL-6, IP-10, and NFκB), and neuroprotective factor (BDNF) were analyzed. Cognitive assessments were performed using RBANS, TMT, TIB and MMSE. POCD patients exhibited an initial upregulation of NRF2-related antioxidant genes, which failed to sustain at 3-months follow-up, leading to a decline in HMOX1, IP-10 and BDNF protein levels, along with increased LOX-1 protein level and NFκB expression, indicating persistent oxidative stress and inflammation. In contrast, non-POCD patients demonstrated a sustained increase in antioxidant and neuroprotective markers, suggesting a more effective compensatory response. ROC analysis identified HMOX1 and BDNF as significant predictors of POCD, with LOX-1 and IP-10 emerging as diagnostic markers at follow-up. In conclusion, our findings highlight the dynamic regulation of oxidative stress and inflammatory pathways in POCD, emphasizing the failure of sustained neuroprotection in affected patients. Further large-scale studies are necessary to validate these findings, and biomarker-based screening could facilitate early risk stratification and targeted interventions to improve cognitive outcomes after cardiac surgery.
Collapse
Affiliation(s)
- Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy.
| | - Emanuele Lo Gerfo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
- NeuroMI - Milan Center for Neuroscience, 20100, Milan, Italy
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), University of Pittsburgh Medical Center (UPMC), 90127, Palermo, Italy
| | - Giovanna Russelli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
| | - Matteo Bulati
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
| | - Rosaria Tinnirello
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
| | - Maura Cimino
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127, Palermo, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, 00161, Rome, Italy
| | - Federica Avorio
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), University of Pittsburgh Medical Center (UPMC), 90127, Palermo, Italy
| | - Vincenzina Lo Re
- Neurology Service, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), University of Pittsburgh Medical Center (UPMC), 90127, Palermo, Italy.
| |
Collapse
|
11
|
Pang Y, Li Q, Sergi Z, Yu G, Kim O, Lu P, Chan M, Sang X, Wang H, Ranjan A, Robey RW, Soheilian F, Tran B, Núñez FJ, Zhang M, Song H, Zhang W, Davis D, Gilbert MR, Gottesman MM, Liu Z, Thomas CJ, Castro MG, Gujral TS, Wu J. Exploiting the therapeutic vulnerability of IDH-mutant gliomas with zotiraciclib. iScience 2025; 28:112283. [PMID: 40241769 PMCID: PMC12001108 DOI: 10.1016/j.isci.2025.112283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 08/07/2024] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Isocitrate dehydrogenase (IDH)-mutant gliomas have distinctive metabolic and biological traits that potentially render them susceptible to targeted treatments. Here, by conducting a high-throughput drug screen, we pinpointed a specific vulnerability of IDH-mutant gliomas to zotiraciclib (ZTR). ZTR exhibited selective growth inhibition across multiple IDH-mutant glioma in vitro and in vivo models. Mechanistically, ZTR at low doses suppressed CDK9 and RNA Pol II phosphorylation in IDH-mutant cells, disrupting mitochondrial function and NAD+ production, resulting in oxidative stress. Integrated biochemical profiling of ZTR kinase targets and transcriptomics unveiled that ZTR-induced bioenergetic failure was linked to the suppression of PIM kinase activity. We posit that the combination of mitochondrial dysfunction and an inability to adapt to oxidative stress resulted in significant cell death upon ZTR treatment, ultimately increasing the therapeutic vulnerability of IDH-mutant gliomas. These findings prompted a clinical trial evaluating ZTR in IDH-mutant gliomas (NCT05588141).
Collapse
Affiliation(s)
- Ying Pang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qi Li
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zach Sergi
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guangyang Yu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olga Kim
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peng Lu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marina Chan
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Xueyu Sang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Herui Wang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alice Ranjan
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert W. Robey
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ferri Soheilian
- Electron Microscopy Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Bao Tran
- Cancer Research Technology Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 20701, USA
| | - Felipe J. Núñez
- Departments of Neurosurgery and Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Meili Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hua Song
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Zhang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dionne Davis
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark R. Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael M. Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhenggang Liu
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Craig J. Thomas
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, Rockville, MD 20850, USA
| | - Maria G. Castro
- Departments of Neurosurgery and Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Taranjit S. Gujral
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jing Wu
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Park SJ, Cerella C, Kang JM, Byun J, Kum D, Orlikova-Boyer B, Lorant A, Schnekenburger M, Al-Mourabit A, Christov C, Lee J, Han BW, Diederich M. Tetrahydrobenzimidazole TMQ0153 targets OPA1 and restores drug sensitivity in AML via ROS-induced mitochondrial metabolic reprogramming. J Exp Clin Cancer Res 2025; 44:114. [PMID: 40197337 PMCID: PMC11974110 DOI: 10.1186/s13046-025-03372-0] [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: 12/01/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly aggressive cancer with a 5-year survival rate of less than 35%. It is characterized by significant drug resistance and abnormal energy metabolism. Mitochondrial dynamics and metabolism are crucial for AML cell survival. Mitochondrial fusion protein optic atrophy (OPA)1 is upregulated in AML patients with adverse mutations and correlates with poor prognosis. METHOD This study investigated targeting OPA1 with TMQ0153, a tetrahydrobenzimidazole derivative, to disrupt mitochondrial metabolism and dynamics as a novel therapeutic approach to overcome treatment resistance. Effects of TMQ0153 treatment on OPA1 and mitofusin (MFN)2 protein levels, mitochondrial morphology, and function in AML cells. In this study, we examined reactive oxygen species (ROS) production, oxidative phosphorylation (OXPHOS) inhibition, mitochondrial membrane potential (MMP) depolarization, and apoptosis. Additionally, metabolic profiling was conducted to analyze changes in metabolic pathways. RESULTS TMQ0153 treatment significantly reduced OPA1 and mitofusin (MFN)2 protein levels and disrupted the mitochondrial morphology and function in AML cells. This increases ROS production and inhibits OXPHOS, MMP depolarization, and caspase-dependent apoptosis. Metabolic reprogramming was observed, shifting from mitochondrial respiration to glycolysis and impaired respiratory chain activity. Profiling revealed reduced overall metabolism along with changes in the glutathione (GSH)/oxidized glutathione (GSSG) and NAD⁺/NADH redox ratios. TMQ0153 treatment reduces tumor volume and weight in MV4-11 xenografts in vivo. Combination therapies with TMQ0153 and other AML drugs significantly reduced the leukemic burden and prolonged survival in NOD scid gamma (NSG) mice xenografted with U937-luc and MOLM-14-luc cells. CONCLUSION TMQ0153 targets mitochondrial dynamics by inhibiting OPA1, inducing metabolic reprogramming, and triggering apoptosis in AML cells. It enhances the efficacy of existing AML therapies and provides a promising combination treatment approach that exploits mitochondrial vulnerability and metabolic reprogramming to improve treatment outcomes in AML.
Collapse
MESH Headings
- Humans
- GTP Phosphohydrolases/metabolism
- GTP Phosphohydrolases/antagonists & inhibitors
- GTP Phosphohydrolases/genetics
- Reactive Oxygen Species/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Animals
- Mice
- Benzimidazoles/pharmacology
- Mitochondria/metabolism
- Mitochondria/drug effects
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Apoptosis/drug effects
- Oxidative Phosphorylation/drug effects
- Drug Resistance, Neoplasm/drug effects
- Membrane Potential, Mitochondrial/drug effects
- Metabolic Reprogramming
Collapse
Affiliation(s)
- Su Jung Park
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Jin Mo Kang
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jinyoung Byun
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - David Kum
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Barbora Orlikova-Boyer
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Anne Lorant
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Luxembourg Centre for Systems Biomedicine, Bioinformatics Core, Roudeneck, 1, Boulevard du Jazz, Esch-sur-Alzette, L-4370, Luxembourg
| | - Michael Schnekenburger
- Laboratoire de Biologie Moléculaire du Cancer, BAM3 Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
- Present address: Department of Cancer Research, Luxembourg Institute of Health (LIH), BAM Pavillon 2, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Ali Al-Mourabit
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-Sur-Yvette, 91190, France
| | - Christo Christov
- Service d'Histologie, Faculté de Médicine, Université de Lorraine, and INSERM U1256 NGERE, 54000, Nancy, France
| | - Juyong Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Medicine, Seoul National University, 1 Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Korea
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Marc Diederich
- Research Institute of Pharmaceutical Sciences & Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
13
|
Ashwani, Sharma A, Choudhary MK, Gugulothu D, Pandita D, Verma S, Vora LK, Khatri DK, Garabadu D. Epigenetic and Mitochondrial Metabolic Dysfunction in Multiple Sclerosis: A Review of Herbal Drug Approaches and Current Clinical Trials. Mol Neurobiol 2025:10.1007/s12035-025-04868-8. [PMID: 40180689 DOI: 10.1007/s12035-025-04868-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disease characterised by inflammation, demyelination, and neurodegeneration within the central nervous system (CNS). While the exact causes remain unclear, recent research highlights the significant role of epigenetic modifications and mitochondrial dysfunction in the disease's onset and progression. Epigenetic alterations, such as DNA methylation, histone modification, and microRNA regulation, influence gene expression without altering the DNA sequence, leading to immune dysregulation and inflammation. Similarly, mitochondrial dysfunction, marked by impaired oxidative phosphorylation, reduced adenosine triphosphate (ATP) production, and increased reactive oxygen species (ROS), contributes to neurodegeneration and impaired remyelination in MS. The growing interest in targeting these two interconnected mechanisms has opened new avenues for MS treatment. Herbal drugs, known for their multi-targeted effects, have shown potential in modulating epigenetic markers and enhancing mitochondrial function. Compounds such as resveratrol, curcumin, epigallocatechin-3-gallate (EGCG), quercetin, and omega-3 fatty acids demonstrate potential in regulating DNA methylation, histone deacetylation, and mitochondrial biogenesis. These natural agents offer dual-action therapies by reducing oxidative stress and inflammation while promoting neuronal survival and remyelination. This review explores the therapeutic potential of herbal drugs targeting epigenetic and mitochondrial pathways in MS, evaluating their mechanisms of action and highlighting their promise as novel therapeutic agents. While initial findings are encouraging, further research and clinical trials are required to validate the efficacy of these herbal treatments and fully understand their potential in slowing disease progression and improving patient outcomes in MS. Such exploration could pave the way for safer, multi-targeted therapies, offering new hope in the management of MS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Ashwani
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | | | - Mayank Kumar Choudhary
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India
| | - Dalapathi Gugulothu
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India.
| | - Deepti Pandita
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Surajpal Verma
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, 110017, India
| | - Lalitkumar K Vora
- School of Pharmacy, Medical Biology Centre, Queen'S University Belfast, 97 Lisburn Road, Belfast, Northern Ireland, BT9 7BL, UK.
| | - Dharmendra Kumar Khatri
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, 303121, India.
| | - Debapriya Garabadu
- Department of Pharmacology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151401, India.
| |
Collapse
|
14
|
Milosavljevic S, Piroli MV, Sandago EJ, Piroli GG, Smith HH, Beggiato S, Frizzell N, Pocivavsek A. Parental kynurenine 3-monooxygenase genotype in mice directs sex-specific behavioral outcomes in offspring. Biol Sex Differ 2025; 16:22. [PMID: 40176166 PMCID: PMC11967062 DOI: 10.1186/s13293-025-00703-w] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Disruptions in brain development can impact behavioral traits and increase the risk of neurodevelopmental conditions such as autism spectrum disorder, attention-deficit/hyperactivity disorder (ADHD), schizophrenia, and bipolar disorder, often in sex-specific ways. Dysregulation of the kynurenine pathway (KP) of tryptophan metabolism has been implicated in cognitive and neurodevelopmental disorders. Increased brain kynurenic acid (KYNA), a neuroactive metabolite implicated in cognition and sleep homeostasis, and variations in the Kmo gene, which encodes kynurenine 3-monooxygenase (KMO), have been identified in these patients. We hypothesize that parental Kmo genetics influence KP biochemistry, sleep behavior and brain energy demands, contributing to impairments in cognition and sleep in offspring through the influence of parental genotype and genetic nurture mechanisms. METHODS A mouse model of partial Kmo deficiency, Kmo heterozygous (HET-Kmo+/-), was used to examine brain KMO activity, KYNA levels, and sleep behavior in HET-Kmo+/- compared to wild-type control (WT-Control) mice. Brain mitochondrial respiration was assessed, and KP metabolites and corticosterone levels were measured in breast milk. Behavioral assessments were conducted on wild-type offspring from two parental groups: (i) WT-Control from WT-Control parents, (ii) wild-type Kmo (WT-Kmo+/+) from Kmo heterozygous parents (HET-Kmo+/-). All mice were C57Bl/6J background strain. Adult female and male offspring underwent behavioral testing for learning, memory, anxiety-like behavior and sleep-wake patterns. RESULTS HET-Kmo+/- mice exhibited reduced brain KMO activity, increased KYNA levels, and disrupted sleep architecture and electroencephalogram (EEG) power spectra. Mitochondrial respiration (Complex I and Complex II activity) and electron transport chain protein levels were impaired in the hippocampus of HET-Kmo+/- females. Breast milk from HET-Kmo+/- mothers increased kynurenine exposure during lactation but corticosterone levels were unchanged. Compared to WT-Control offspring, WT-Kmo+/+ females showed impaired spatial learning, heightened anxiety, reduced sleep and abnormal EEG spectral power. WT-Kmo+/+ males had deficits in reversal learning but no sleep disturbances or anxiety-like behaviors. CONCLUSIONS These findings suggest that Kmo deficiency impacts KP biochemistry, sleep behavior, and brain mitochondrial function. Even though WT-Kmo+/+ inherit identical genetic material as WT-Control, their development might be shaped by the parent's physiology, behavior, or metabolic state influenced by their Kmo genotype, leading to phenotypic sex-specific differences in offspring.
Collapse
Affiliation(s)
- Snezana Milosavljevic
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Maria V Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Emma J Sandago
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Gerardo G Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Holland H Smith
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA.
| |
Collapse
|
15
|
Singh J, Sah B, Deng Y, Clarke R, Liu L. Molecular mechanisms underlying TXNIP's anti-tumor role in breast cancer, including interaction with a novel, pro-tumor partner: CAST. Cell Death Dis 2025; 16:236. [PMID: 40175348 PMCID: PMC11965567 DOI: 10.1038/s41419-025-07566-4] [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: 08/30/2024] [Revised: 02/22/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Thioredoxin-interacting protein (TXNIP) plays a pivotal role in glucose metabolism and redox signaling. Its emerging function as a potent suppressor of cell proliferation in various cancer contexts underscores its importance in cancer development. In a previous study, we found TXNIP activation by UNC0642, an inhibitor of histone methyltransferase G9A, significantly inhibited MDA-MB-231 breast cancer cell proliferation in vitro and tumor growth in vivo. Here, we demonstrated that TXNIP knockdown increased MDA-MB-231 tumor growth and metastasis in a mouse model. Reintroducing TXNIP into TXNIP-deficient HCC-1954 breast cancer cells decreased cell proliferation and migration while boosting the generation of reactive oxygen species, alongside reductions in mitochondrial respiration, mitochondrial membrane potential, and glycolysis. To elucidate the mechanisms underlying TXNIP's antitumor effects in breast cancer cells, we conducted co-immunoprecipitation and proteomic analyses that revealed calpastatin (CAST) as a novel TXNIP-interacting protein in MDA-MB-231 and HCC-1954 cells. Overexpression of CAST, an endogenous inhibitor of calpains, significantly increased xenograft tumor growth for both MDA-MB-231 and HCC-1954 cells, underscoring its novel role as a tumor promoter. In addition, we identified a positive correlation between the expression of TXNIP and interleukin-24 (IL-24), a molecule that induces cancer-specific apoptosis in several breast cancer cell lines. Our findings also show TXNIP's ability to decrease activation of STAT3, a key driver of oncogenesis. Finally, cells with high levels of TXNIP expression displayed increased susceptibility to IL-24 and WP1066, a specific STAT3 inhibitor, suggesting possible predictive value for TXNIP. Collectively, these findings unveil novel TXNIP-dependent pathways that may contribute to breast cancer pathogenesis, enriching our understanding of this molecule's intricate role in cancer and potentially paving the way for clinical translation.
Collapse
Affiliation(s)
- Jasvinder Singh
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Bindeshwar Sah
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Yibin Deng
- Department of Urology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Biochemistry, Molecular Biology and Biophysics, Minneapolis, MN, 55455, USA
| | - Liang Liu
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Das A, Mitra A, Sarkar S, Ghosh S, Bandyopadhyay D, Chattopadhyay S. Arsenic unsettles the cerebellar balance between neurodegeneration and neurogenesis: reversal by folic acid. Apoptosis 2025; 30:710-733. [PMID: 39720976 DOI: 10.1007/s10495-024-02054-0] [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] [Accepted: 11/30/2024] [Indexed: 12/26/2024]
Abstract
Arsenic-mediated neurodegenerative disorders affect millions of individuals globally, but the specific impact of environmental arsenic on adult cerebellar degeneration and neurogenesis is incompletely understood. Of particular concern is arsenic-induced apoptosis-driven neurodegeneration. Our major objective was to investigate the molecular signaling intricacies associated with arsenic-induced death of cerebellar neurons and to propose folic acid as a possible intervention. Swiss albino mice were treated with sodium arsenite (orally: 0.05 mg/L) and folic acid (orally:10 mg/kg) for 28 days. We observed that arsenic caused noticeable cell loss with morphological alterations in cerebellum, which was remarkably restored by folic acid. Arsenic-induced morphological alterations consequently perturbed transcriptional activities of neural stem cell factors-SOX2 and KLF9, which resulted in the suppression of pro-neurogenic mediators NeuroD1, Neurogenin2, calbindin and NeuN. Interestingly, folic acid reversed the expression of these critical pro-neurogenic mediators to mitigate these degenerative changes to promote neurogenesis. Delving deep, we found that folic acid rescued arsenic-exposed cerebellum from severe oxidative and pro-inflammatory insults by increasing antioxidants like SOD, Catalase, GSH, upregulating Nrf2 and downregulating M1 macrophages, JNK, NF-κB, and STAT3 activities. For the first time, we are reporting that arsenic induced a G1/S cell cycle arrest and triggered apoptosis in mouse cerebellum by activating the p53-p21 axis, downregulating CDKs and instigated p21-mediated suppression of SOX2 transcriptional activity. Folic acid abated such alterations by modulating the p53/p21/SOX2 axis. Collectively, the anti-apoptotic and pro-neurogenic effects of folic acid present it as a promising therapeutic candidate, warranting further research into its efficacy against metal-induced neurodegenerative disorders.
Collapse
Affiliation(s)
- Ankur Das
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
- Department of Physiology, Trivenidevi Bhalotia College, Kazi Nazrul University, Raniganj, West Bengal, 713347, India
| | - Ankan Mitra
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Swaimanti Sarkar
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Sourav Ghosh
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
- Department of Physiology, Ananda Chandra College, University of North Bengal, Jalpaiguri, West Bengal, 735101, India
| | | | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India.
- Centre for Research in Nanoscience and Nanotechnology (CRNN), University of Calcutta, JD-2, Salt Lake, Sector III, Kolkata, 700098, India.
- UCSTA, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, India.
| |
Collapse
|
17
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [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: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
18
|
Srivastava SP, Kopasz-Gemmen O, Thurman A, Rajendran BK, Selvam MM, Kumar S, Srivastava R, Suresh MX, Kumari R, Goodwin JE, Inoki K. The molecular determinants regulating redox signaling in diabetic endothelial cells. Front Pharmacol 2025; 16:1563047. [PMID: 40290438 PMCID: PMC12023289 DOI: 10.3389/fphar.2025.1563047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidation and reduction are vital for keeping life through several prime mechanisms, including respiration, metabolism, and other energy supplies. Mitochondria are considered the cell's powerhouse and use nutrients to produce redox potential and generate ATP and H2O through the process of oxidative phosphorylation by operating electron transfer and proton pumping. Simultaneously, mitochondria also produce oxygen free radicals, called superoxide (O2 -), non-enzymatically, which interacts with other moieties and generate reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), peroxynitrite (ONOO-), and hydroxyl radical (OH-). These reactive oxygen species modify nucleic acids, proteins, and carbohydrates and ultimately cause damage to organs. The nutrient-sensing kinases, such as AMPK and mTOR, function as a key regulator of cellular ROS levels, as loss of AMPK or aberrant activation of mTOR signaling causes ROS production and compromises the cell's oxidant status, resulting in various cellular injuries. The increased ROS not only directly damages DNA, proteins, and lipids but also alters cellular signaling pathways, such as the activation of MAPK or PI3K, the accumulation of HIF-1α in the nucleus, and NFkB-mediated transcription of pro-inflammatory cytokines. These factors cause mesenchymal activation in renal endothelial cells. Here, we discuss the biology of redox signaling that underlies the pathophysiology of diabetic renal endothelial cells.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | | | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Barani Kumar Rajendran
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - M. Masilamani Selvam
- Department of Pharmaceutical Technology, Paavai Engineering College, Namakkal, Tamil Nadu, India
| | - Sandeep Kumar
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Rohit Srivastava
- Laboratory of Medical Transcriptomics, Department of Endocrinology, Nephrology Services, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - M. Xavier Suresh
- School of Advanced Sciences and Languages, VIT Bhopal University, Sehore, Madhya Pradesh, India
| | - Reena Kumari
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
20
|
Adnan M, Siddiqui AJ, Bardakci F, Surti M, Badraoui R, Patel M. Mechanistic Insights into the Neuroprotective Potential of Aegle marmelos (L.) Correa Fruits against Aβ-Induced Cell Toxicity in Human Neuroblastoma SH-SY5Y Cells. Pharmaceuticals (Basel) 2025; 18:489. [PMID: 40283926 PMCID: PMC12030591 DOI: 10.3390/ph18040489] [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: 02/27/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Amyloid-β (Aβ) plaque accumulation, oxidative stress, and cholinergic dysfunction are hallmarks of Alzheimer's disease (AD), a neurodegenerative disability that progresses over time, ultimately resulting in the loss of neurons. The side effects and limitations of current synthetic drugs have shifted attention toward natural alternatives. This study investigates the ethanolic extract of Aegle marmelos (L.) Corrêa fruits for their antioxidant, AChE-inhibitory, and anti-amyloidogenic properties, as well as their neuroprotective effects against amyloid beta-peptide (Aβ1-42). Methods: Phytochemical constituents were identified through HR-LCMS analysis and their antioxidant (DPPH, FRAP) and neuroprotective activities (AChE inhibition, ThT binding, MTT assay, ROS reduction, MMP restoration, and AD-related gene expression via qRT-PCR) were assessed using SHSY-5Y neuroblastoma cells. Results: The extract revealed the existence of flavonoids, phenols, and other bioactive substances. In vitro assays demonstrated strong antioxidant and AChE-inhibitory activities, while the ThT binding assay showed protection against amyloid-β aggregation. The extract exhibited no cytotoxicity in SHSY-5Y cells, even at a concentration of 500 μg/mL, whereas Aβ1-42 at 20 μM induced significant cytotoxicity. Co-treatment with Aβ1-42 (10 μM and 20 μM) and the extract improved cell viability (˃50%) and reduced ROS levels. Additionally, the extract restored mitochondrial membrane potential in Aβ1-42 treated cells, highlighting its role in preserving mitochondrial function. Conclusions: These findings suggest that A. marmelos fruits serve as a powerful source of natural antioxidants, AChE inhibitors, and anti-amyloidogenic agents, positioning them as a compelling option for AD treatment.
Collapse
Affiliation(s)
- Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
- King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Fevzi Bardakci
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Malvi Surti
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| | - Riadh Badraoui
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (M.A.)
| | - Mitesh Patel
- Research and Development Cell (RDC), Parul University, Waghodia, Vadodara 391760, Gujarat, India
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Waghodia, Vadodara 391760, Gujarat, India
| |
Collapse
|
21
|
Mark JR, Tansey MG. Immune cell metabolic dysfunction in Parkinson's disease. Mol Neurodegener 2025; 20:36. [PMID: 40128809 PMCID: PMC11934562 DOI: 10.1186/s13024-025-00827-y] [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: 12/12/2024] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Parkinson's disease (PD) is a multi-system disorder characterized histopathologically by degeneration of dopaminergic neurons in the substantia nigra pars compacta. While the etiology of PD remains multifactorial and complex, growing evidence suggests that cellular metabolic dysfunction is a critical driver of neuronal death. Defects in cellular metabolism related to energy production, oxidative stress, metabolic organelle health, and protein homeostasis have been reported in both neurons and immune cells in PD. We propose that these factors act synergistically in immune cells to drive aberrant inflammation in both the CNS and the periphery in PD, contributing to a hostile inflammatory environment which renders certain subsets of neurons vulnerable to degeneration. This review highlights the overlap between established neuronal metabolic deficits in PD with emerging findings in central and peripheral immune cells. By discussing the rapidly expanding literature on immunometabolic dysfunction in PD, we aim to draw attention to potential biomarkers and facilitate future development of immunomodulatory strategies to prevent or delay the progression of PD.
Collapse
Affiliation(s)
- Julian R Mark
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, 32610, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
- Department of Neurology and Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, 32608, USA.
| |
Collapse
|
22
|
Nahar L, Charoensup R, Kalieva K, Habibi E, Guo M, Wang D, Kvasnica M, Onder A, Sarker SD. Natural products in neurodegenerative diseases: recent advances and future outlook. Front Pharmacol 2025; 16:1529194. [PMID: 40176910 PMCID: PMC11961910 DOI: 10.3389/fphar.2025.1529194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025] Open
Abstract
Neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's are on the rise and pose significant challenges due to the lack of effective treatments. This review critically examines the neuroprotective effects of various natural products derived from plants, marine organisms, and fungi. Natural products have long been used in traditional medicine and are gaining attention in modern drug discovery for their unique properties. The review explains how these natural products can protect neurons by influencing the key biological pathways involved in neurodegeneration. It discusses mechanisms including antioxidant effects, anti-inflammatory actions, modulation of cellular signalling, and support for mitochondrial function. A systematic literature search was conducted to minimize bias and ensure rigorous study selection. Preclinical studies using animal models and cell cultures show that secondary metabolites like polyphenols, alkaloids, and terpenoids can significantly reduce neuronal damage. Some clinical trials have shown promising results. However, challenges such as bioavailability, standardization, and dosage must be addressed to translate these findings into clinical practice. The review also evaluates the potential synergy of combining natural products with conventional treatments, offering a complementary therapeutic approach. Natural products represent a promising avenue for developing innovative treatments for neurodegenerative diseases. The review highlights key research gaps and proposes future directions. Future studies should focus on overcoming existing challenges and refining these natural products to improve their efficacy and safety in clinical settings. The application of existing knowledge has the potential to significantly enhance the quality of life for individuals affected by neurodegenerative diseases.
Collapse
Affiliation(s)
- L. Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czechia
| | - R. Charoensup
- School of Integrative Medicine and Medicinal Plants Innovation Center of Mae Fah Luang University, Chiang Rai, Thailand
| | - Kulyash Kalieva
- Department of Chemistry and Mathematics, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - E. Habibi
- Department of Pharmacognosy, Faculty of Pharmacy, Medicinal Plants Research Centre, Mazandaran University of Medical Sciences, Sari, Iran
| | - M. Guo
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, China
| | - D. Wang
- International Joint Laboratory of Medicinal Food Development and Health Products Creation, Biological Engineering Technology Innovation Center of Shandong Province, Heze Branch of Qilu University of Technology (Shandong Academy of Sciences), Heze, China
| | - M. Kvasnica
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czechia
| | - A. Onder
- Department of Pharmacognosy, Faculty of Pharmacy, Ankara University, Ankara, Türkiye
| | - S. D. Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
23
|
Al Tahan MA, Marwah MK, Dhaliwal M, Diaz Sanchez L, Shokr H, Kaur M, Ahmad S, Badhan RKS, Dias IHK, Sanchez-Aranguren L. Novel AP39-Loaded Liposomes Sustain the Release of Hydrogen Sulphide, Enhance Blood-Brain Barrier Permeation, and Abrogate Oxidative Stress-Induced Mitochondrial Dysfunction in Brain Cells. Drug Des Devel Ther 2025; 19:2067-2079. [PMID: 40124553 PMCID: PMC11930254 DOI: 10.2147/dddt.s507697] [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: 12/15/2024] [Accepted: 03/08/2025] [Indexed: 03/25/2025] Open
Abstract
Background Neurodegenerative diseases are often linked to oxidative stress (OS), which worsen neuroinflammation and cause neuronal damage. Managing OS with gasotransmitters such as hydrogen sulphide (H2S) is a promising therapeutic approach to protecting brain cells from oxidative damage. AP39, a mitochondria-targeted H2S donor, has shown neuroprotective potential by reducing OS and improving mitochondrial function. However, its clinical application is limited due to poor stability and rapid release, necessitating a drug delivery system to enhance therapeutic efficacy. Purpose This study aimed to develop a novel AP39-loaded liposomal formulation to provide controlled H2S release, facilitate AP39 permeation across the blood-brain barrier (BBB), and assess functional effects in mitigating oxidative stress and preserving mitochondrial function. Methods AP39-loaded unilamellar liposomes were prepared via ethanol injection and characterised for size, polydispersity, and zeta potential. Entrapment efficiency was determined using HPLC, while cytotoxicity was assessed in human vein endothelial (HUVEC) and neuroblastoma (SHSY5Y) cells. Liposomal permeability, AP39 release kinetics, and cellular uptake were evaluated using a microvasculature BBB model. Mitochondrial function under oxidative stress was assessed using a Seahorse XFe24 Analyzer. Results AP39-loaded liposomes had an average size of 135.92 ± 10.05 nm, a zeta potential of 17.35 ± 3.40 mV, and an entrapment efficiency of 84.48% ± 4.7. Cytotoxicity studies showed no adverse effects after 4 h. Cellular uptake of encapsulated AP39 was significantly higher (7.13 ± 0.28 µg) than the free form (5.8 ± 0.31 µg). The BBB model demonstrated sustained AP39 release (7.28 µg/mL vs 6.44 µg/mL for free AP39). Mitochondrial assays confirmed liposomal AP39 preserved H2S antioxidant properties and enhanced oxygen consumption. Conclusion Our novel liposomal formulation encapsulating AP39 improves stability, promotes sustained release, and enhances BBB permeability while preserving antioxidant effects. These findings indicate that liposomal AP39 is a suitable therapeutic approach to further investigate in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | - Hala Shokr
- Pharmacy Division, University of Manchester, Manchester, UK
| | - Manjit Kaur
- School of Health and Care, Coventry University, Coventry, UK
| | - Shakil Ahmad
- Aston Medical School, Aston University, Birmingham, UK
| | | | | | | |
Collapse
|
24
|
Golpour-Hamedani S, Bagherniya M, Khorvash F, Feizi A, Sharma M, Askari G. The effects of concurrent alpha-linolenic acid, L-carnitine supplementation on clinical symptoms, mental health, and quality of life in women with migraine: a randomized, triple-blind, placebo-controlled trial. Nutr J 2025; 24:40. [PMID: 40082970 PMCID: PMC11905556 DOI: 10.1186/s12937-025-01107-7] [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: 11/15/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Migraine, as a widespread neurological condition, substantially impacts quality of life, particularly among women. Therefore, this study aimed to explore the potential effects of alpha-linolenic acid (ALA) and L-carnitine co-supplementation on migraine symptoms, mental health, and life quality in women with migraine. METHODS In this randomized, triple-blind, placebo-controlled trial, 80 women with migraine were randomly assigned to receive either ALA (1000 mg) plus L-carnitine (500 mg) or matching placebos daily for 12 weeks. Migraine characteristics, mental health parameters, and quality of life measures were assessed at baseline and study end. RESULTS The intervention group demonstrated a significant reduction in migraine frequency (-2.96; 95% CI (-3.48, -2.45) vs -0.07; 95% CI (-0.68, 0.53), P < 0.001), severity (-1.6; 95% CI (-2.05, -1.15) vs - 0.44; 95% CI (-0.91, 0.02), P = 0.001), and duration (-4.9; 95% CI (-6.34, -3.45) vs -0.5; 95% CI (-1.06, 0.66) hours, P < 0.001) compared to the placebo group. Mental health improvements were observed in depression (-7.4; 95% CI (-9.24, -5.55) vs 0.05; 95% CI (-1.16, 1.26), P < 0.001), and anxiety scores (-5.7; 95% CI (-7.26, -4.14) vs - 0.65; 95% CI (-2.33, 1.03), P < 0.001). Quality of life measures showed significant enhancement, with increased migraine-specific quality of life (9.75; 95% CI (8.01, 11.49) vs 1.22; 95% CI (-0.66, 3.11), P < 0.001) and decreased headache impact test-6 scores (-8.57; 95% CI (-11.79, -5.36) vs -1.35; 95% CI (-3.41, 0.71), P = 0.005) in the intervention group compared to the controls. CONCLUSION Co-supplementation with ALA and L-carnitine may offer a promising adjuvant therapy for managing migraine in women, addressing both physical symptoms and psychological burdens. TRIAL REGISTRATION IRCT20121216011763N57.
Collapse
Affiliation(s)
- Sahar Golpour-Hamedani
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fariborz Khorvash
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Department of Epidemiology and Biostatistics, School of Health, Clinical Toxicology Research Center and Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Manoj Sharma
- Social and Behavioral Health, School of Public Health, University of Nevada, Las Vegas, NV, USA
- Department of Internal Medicine, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas, NV, USA
| | - Gholamreza Askari
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
25
|
Salimi A, Shabani M, Shahsavar SP, Naserian A, Khezri S, Karroubian H. Mitochondrial transplantation via injection of exogenous mitochondria into blood reduces bleomycin-induced oxidative damages and mitochondrial dysfunction in lung tissue. J Mol Histol 2025; 56:104. [PMID: 40063258 DOI: 10.1007/s10735-025-10386-7] [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: 01/01/2025] [Accepted: 02/27/2025] [Indexed: 04/25/2025]
Abstract
Mechanistic studies have been suggested that adverse effect of bleomycin is attributed to formation of free radicals, mitochondria damages, oxidative stress and inflammation in lung tissue. Mitochondria act as central regulators in the oxidative stress and inflammatory responses in lung tissue, then it can be a promising approach for management bleomycin-induced pneumotoxicity. In the current study, we aim to investigated the injection of exogenous mitochondria into blood as one of the most promising pharmacological approaches to reduce bleomycin-induced lung toxicity in rats. Rats were divided into 4 groups as control, bleomycin (5 mg/kg), bleomycin + mitochondria (250 µg/kg), and mitochondria (250 µg/kg) alone. After 2 weeks, the survival rate, weight changes of animals, wet/dry ratio of lung tissue, alterations of histopathology, hydroxyproline content, oxidative stress and mitochondrial biomarkers were determined. Except the survival rate, weight changes of animals and wet/dry ratio of lung tissue, administration of bleomycin resulted in significant alteration in GSH content, MDA level, hydroxyproline amount, collapse of mitochondrial membrane potential (MMP), reduction of succinate dehydrogenases (SDH) activity and histopathological abnormality in comparison with control group. While exogenous mitochondria could inhibit GSH depletion, reduce production of MDA, improve the activity of SDH, prevent loss of MMP and histopathological abnormality. To the best of our knowledge, our data provides the first direct experimental evidence that injection of exogenous mitochondria into blood is capable of ameliorating bleomycin-induced lung toxicity in rats. These findings support that mitochondrial transplantation can be a promising therapeutic strategy for bleomycin-associated mitochondrial dysfunction and lung damage.
Collapse
Affiliation(s)
- Ahmad Salimi
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, P.O. Box: 56189-53141, Ardabil, Iran.
| | - Mohammad Shabani
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, P.O. Box: 56189-53141, Ardabil, Iran
| | | | - Aida Naserian
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Saleh Khezri
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, P.O. Box: 56189-53141, Ardabil, Iran
| | - Hamed Karroubian
- Students Research Committee, Faculty of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
26
|
Ivanov MV, Kopeykina AS, Kazakova EM, Tarasova IA, Sun Z, Postoenko VI, Yang J, Gorshkov MV. Modified Decision Tree with Custom Splitting Logic Improves Generalization across Multiple Brains' Proteomic Data Sets of Alzheimer's Disease. J Proteome Res 2025; 24:1053-1066. [PMID: 39984290 DOI: 10.1021/acs.jproteome.4c00677] [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: 02/23/2025]
Abstract
Many factors negatively affect a generalization of the findings in discovery proteomics. They include differentiation between patient cohorts, a variety of experimental conditions, etc. We presented a machine-learning-based workflow for proteomics data analysis, aiming at improving generalizability across multiple data sets. In particular, we customized the decision tree model by introducing a new parameter, min_groups_leaf, which regulates the presence of the samples from each data set inside the model's leaves. Further, we analyzed a trend for the feature importance's curve as a function of the novel parameter for feature selection to a list of proteins with significantly improved generalization. The developed workflow was tested using five proteomic data sets obtained for post-mortem human brain samples of Alzheimer's disease. The data sets consisted of 535 LC-MS/MS acquisition files. The results were obtained for two different pipelines of data processing: (1) MS1-only processing based on DirectMS1 search engine and (2) a standard MS/MS-based one. Using the developed workflow, we found seven proteins with expression patterns that were unique for asymptomatic Alzheimer patients. Two of them, Serotransferrin TRFE and DNA repair nuclease APEX1, may be potentially important for explaining the lack of dementia in patients with the presence of neuritic plaques and neurofibrillary tangles.
Collapse
Affiliation(s)
- Mark V Ivanov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Anna S Kopeykina
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Elizaveta M Kazakova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Irina A Tarasova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Zhao Sun
- Clinical Systems Biology Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou 450052, China
| | - Valeriy I Postoenko
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Jinghua Yang
- Clinical Systems Biology Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou 450052, China
| | - Mikhail V Gorshkov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
27
|
Ngcobo NN. Influence of Ageing on the Pharmacodynamics and Pharmacokinetics of Chronically Administered Medicines in Geriatric Patients: A Review. Clin Pharmacokinet 2025; 64:335-367. [PMID: 39798015 PMCID: PMC11954733 DOI: 10.1007/s40262-024-01466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2024] [Indexed: 01/13/2025]
Abstract
As people age, the efficiency of various regulatory processes that ensure proper communication between cells and organs tends to decline. This deterioration can lead to difficulties in maintaining homeostasis during physiological stress. This includes but is not limited to cognitive impairments, functional difficulties, and issues related to caregivers which contribute significantly to medication errors and non-adherence. These factors can lead to higher morbidity, extended hospital stays, reduced quality of life, and even mortality. The decrease in homeostatic capacity varies among individuals, contributing to the greater variability observed in geriatric populations. Significant pharmacokinetic and pharmacodynamic alterations accompany ageing. Pharmacokinetic changes include decreased renal and hepatic clearance and an increased volume of distribution for lipid-soluble drugs, which prolong their elimination half-life. Pharmacodynamic changes typically involve increased sensitivity to various drug classes, such as anticoagulants, antidiabetic and psychotropic medications. This review examines the primary age-related physiological changes in geriatrics and their impact on the pharmacokinetics and pharmacodynamics of medications.
Collapse
Affiliation(s)
- Nokwanda N Ngcobo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
28
|
Nigdelioglu Dolanbay S, Şirin S, Aslim B. Allocryptopine Attenuates Inflammatory Responses in Microglial Cells Via TLR4-Dependent NF-κB and p38 MAPK Pathways. Mol Neurobiol 2025; 62:3833-3847. [PMID: 39331354 DOI: 10.1007/s12035-024-04520-x] [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: 05/14/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Studies in the existing literature propose that allocryptopine possesses both antioxidant and anti-inflammatory properties, showcasing its neuroprotective effects by potentially mitigating oxidative stress and inflammation. This study aims to investigate the antioxidant and anti-inflammatory effects of allocryptopine on various targets and potential mechanisms that have not been previously explored in the literature. Initially, we used MTT and LDH methods to evaluate the effects of allocryptopine on cell viability in BV-2 cells exposed to LPS-induced damage. Subsequently, we evaluated the impact of allocryptopine on pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), other inflammatory mediators (Cox-2 and iNOS), and p38 MAPK genes and proteins through qRT-PCR and Western blot analyses. Also, we evaluated the impact of allocryptopine on NF-κB proteins (TLR4, MyD88, IκBα, p-p50, and p-p65) through ELISA assay. Molecular docking analyses were performed to investigate the potential binding of allocryptopine to target proteins (TLR4, MyD88, IκBα, p50, p65, MKK3, MKK4, MKK6, p38, AP-1 (c-Jun and ATF2), IL-1β, IL-6, TNF-α, Cox-2, and iNOS) associated with the TLR4, NF-κB, and p38 MAPK pathways. Our results indicate that allocryptopine exerts a comprehensive influence on pro-inflammatory cytokines and other inflammatory mediators by inhibiting TLR4 signaling and modulating the NF-κB and p38 MAPK pathways. The outcomes of our study suggest that the antioxidant and anti-inflammatory efficacy of allocryptopine is intricately linked to the modulation of key molecular pathways associated with oxidative stress and inflammation. These findings highlight the potential of allocryptopine as a therapeutic agent for addressing neurodegenerative diseases by safeguarding neuronal health.
Collapse
Affiliation(s)
| | - Seda Şirin
- Faculty of Science, Department of Biology, Gazi University, 06500, Teknikokullar, Ankara, Turkey.
| | - Belma Aslim
- Faculty of Science, Department of Biology, Gazi University, 06500, Teknikokullar, Ankara, Turkey
| |
Collapse
|
29
|
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
|
30
|
Kong L, Li S, Fu Y, Cai Q, Du X, Liang J, Ma T. Mitophagy in relation to chronic inflammation/ROS in aging. Mol Cell Biochem 2025; 480:721-731. [PMID: 38834837 DOI: 10.1007/s11010-024-05042-9] [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: 04/07/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
Various assaults on mitochondria occur during the human aging process, contributing to mitochondrial dysfunction. This mitochondrial dysfunction is intricately connected with aging and diseases associated with it. In vivo, the accumulation of defective mitochondria can precipitate inflammatory and oxidative stress, thereby accelerating aging. Mitophagy, an essential selective autophagy process, plays a crucial role in managing mitochondrial quality control and homeostasis. It is a highly specialized mechanism that systematically removes damaged or impaired mitochondria from cells, ensuring their optimal functioning and survival. By engaging in mitophagy, cells are able to maintain a balanced and stable environment, free from the potentially harmful effects of dysfunctional mitochondria. An ever-growing body of research highlights the significance of mitophagy in both aging and age-related diseases. Nonetheless, the association between mitophagy and inflammation or oxidative stress induced by mitochondrial dysfunction remains ambiguous. We review the fundamental mechanisms of mitophagy in this paper, delve into its relationship with age-related stress, and propose suggestions for future research directions.
Collapse
Affiliation(s)
- Liang Kong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Shuhao Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Yu Fu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Qinyun Cai
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Xinyun Du
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
31
|
Piccoli M, Barbato L, Maiorana NV, Mingione A, Raimondo F, Ghirimoldi M, Cirillo F, Schiepati M, Salerno D, Anastasia L, Albi E, Manfredi M, Bocci T, Priori A, Signorelli P. Direct Current Stimulation (DCS) Modulates Lipid Metabolism and Intercellular Vesicular Trafficking in SHSY-5Y Cell Line: Implications for Parkinson's Disease. J Neurochem 2025; 169:e70014. [PMID: 39930930 PMCID: PMC11811683 DOI: 10.1111/jnc.70014] [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: 10/04/2024] [Revised: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
The modulation of the brain's electrical activity for therapeutic purposes has recently gained attention, supported by the promising results obtained through the non-invasive application of transcranial direct current stimulation (tDCS) in the treatment of neurodegenerative and neurological diseases. To optimize therapeutic efficacy, it is crucial to investigate the cellular and molecular effects of tDCS. This will help to identify important biomarkers, predict patient's response and develop personalized treatments. In this study, we applied direct current stimulation (DCS) to a neural cell line, using mild currents over short periods of time (0.5 mA, 20 min), with 24-h intervals. We observed that DCS induced changes in the cellular lipidome, with transient effects observed after a single stimulation (lasting 24 h) and more significant, long-lasting effects (up to 72 h) after repeated stimulation cycles. In neural cells, multiple DCS treatment modulated structural membrane lipids (PE, PS, PI), downregulated glycerol lipids with ether-linked fatty acids and pro-inflammatory lipids (ceramides and lyso-glycerophospholipids) (p ≤ 0.005). Multiple DCS sessions altered transcriptional activity by decreasing the expression of inflammatory cytokines (TNF-α, p ≤ 0.05; IL-1β, p ≤ 0.01), while increasing the expression of neuroprotective factors such as heme oxygenase-1 (p ≤ 0.0001) and brain-derived neurotrophic factor (p ≤ 0.05), as well as proteins involved in vesicular transport (SNARE, sorting nexins and seipin and α-synuclein; p ≤ 0.05). In addition, DCS enhanced the release of extracellular vesicles, with repeated stimulations significantly increasing the release of exosomes threefold. In conclusion, while a single electrical stimulation induces transient metabolic changes with limited phenotypic effects, repeated applications induce a broader and deeper modulation of lipid species. This may lead to a neuroprotective and neuroplasticity-focussed transcriptional profile, potentially supporting the therapeutic effects of tDCS at the cellular and molecular level in patients..
Collapse
Affiliation(s)
- Marco Piccoli
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- School of MedicineUniversity Vita‐Salute San RaffaeleMilanItaly
| | - Luisa Barbato
- Biochemistry LaboratoryIRCCS Policlinico San DonatoMilanItaly
| | | | - Alessandra Mingione
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | | | - Marco Ghirimoldi
- Biological Mass Spectrometry Lab, Department of Translational MedicineUniversity of Piemonte OrientaleNovaraItaly
| | - Federica Cirillo
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
| | - Mattia Schiepati
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Domenico Salerno
- School of Medicine and Surgery BioNanoMedicine Center NANOMIBUniversity of Milan‐BicoccaMonzaItaly
| | - Luigi Anastasia
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- School of MedicineUniversity Vita‐Salute San RaffaeleMilanItaly
| | - Elisabetta Albi
- Department of Pharmaceutical Sciences, Interno Orto BotanicoUniversity of PerugiaPerugiaItaly
| | - Marcello Manfredi
- Institute for Molecular and Translational Cardiology (IMTC)IRCCS Policlinico San DonatoMilanItaly
- Biological Mass Spectrometry Lab, Department of Translational MedicineUniversity of Piemonte OrientaleNovaraItaly
- Center for Translational Research Autoimmune Diseases and Allergic DiseasesUniversity of Piemonte OrientaleNovaraItaly
| | - Tommaso Bocci
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Alberto Priori
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| | - Paola Signorelli
- Biochemistry LaboratoryIRCCS Policlinico San DonatoMilanItaly
- “Aldo Ravelli” Research Centre, Department of Health SciencesUniversity of MilanMilanItaly
| |
Collapse
|
32
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
33
|
Wang J, Liu M, Zhao J, Hu P, Gao L, Tian S, Zhang J, Liu H, Xu X, He Z. Oxidative stress and dysregulated long noncoding RNAs in the pathogenesis of Parkinson's disease. Biol Res 2025; 58:7. [PMID: 39871377 PMCID: PMC11770960 DOI: 10.1186/s40659-025-00585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 01/07/2025] [Indexed: 01/29/2025] Open
Abstract
Parkinson's disease (PD) is a progressive age-related neurodegenerative disease whose annual incidence is increasing as populations continue to age. Although its pathogenesis has not been fully elucidated, oxidative stress has been shown to play an important role in promoting the occurrence and development of the disease. Long noncoding RNAs (lncRNAs), which are more than 200 nucleotides in length, are also involved in the pathogenesis of PD at the transcriptional level via epigenetic regulation, or at the post-transcriptional level by participating in physiological processes, including aggregation of the α-synuclein, mitochondrial dysfunction, oxidative stress, calcium stabilization, and neuroinflammation. LncRNAs and oxidative stress are correlated during neurodegenerative processes: oxidative stress affects the expression of multiple lncRNAs, while lncRNAs regulate many genes involved in oxidative stress responses. Oxidative stress and lncRNAs also affect other processes associated with neurodegeneration, including mitochondrial dysfunction and increased neuroinflammation that lead to neuronal death. Therefore, modulating the levels of specific lncRNAs may alleviate pathological oxidative damage and have neuroprotective effects. This review discusses the general mechanisms of oxidative stress, pathological mechanism underlying the role of oxidative stress in the pathogenesis of PD, and teases out the mechanisms through which lncRNAs regulate oxidative stress during PD pathogenesis, as well as identifies the possible neuroprotective mechanisms of lncRNAs. Reviewing published studies will help us further understand the mechanisms underlying the role of lncRNAs in the oxidative stress process in PD and to identify potential therapeutic strategies for PD.
Collapse
Affiliation(s)
- Jialu Wang
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Meitong Liu
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jiuhan Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Pan Hu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Lianbo Gao
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Shen Tian
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Jin Zhang
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China
| | - Huayan Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xiaoxue Xu
- Department of Neurology, First Affiliated Hospital of China Medical University, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
- Key Laboratory of Neurological Disease Big Data of Liaoning Province, No.155 Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, China.
| | - Zhenwei He
- Department of Neurology, Fourth Affiliated Hospital of China Medical University, No.4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
34
|
Chandimali N, Bak SG, Park EH, Lim HJ, Won YS, Kim EK, Park SI, Lee SJ. Free radicals and their impact on health and antioxidant defenses: a review. Cell Death Discov 2025; 11:19. [PMID: 39856066 PMCID: PMC11760946 DOI: 10.1038/s41420-024-02278-8] [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: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Free radicals, characterized by the presence of unpaired electrons, are highly reactive species that play a significant role in human health. These molecules can be generated through various endogenous processes, such as mitochondrial respiration and immune cell activation, as well as exogenous sources, including radiation, pollution, and smoking. While free radicals are essential for certain physiological processes, such as cell signaling and immune defense, their overproduction can disrupt the delicate balance between oxidants and antioxidants, leading to oxidative stress. Oxidative stress results in the damage of critical biomolecules like DNA, proteins, and lipids, contributing to the pathogenesis of various diseases. Chronic conditions such as cancer, cardiovascular diseases, neurodegenerative disorders, and inflammatory diseases have been strongly associated with the harmful effects of free radicals. This review provides a comprehensive overview of the characteristics and types of free radicals, their mechanisms of formation, and biological impacts. Additionally, we explore natural compounds and extracts studied for their antioxidant properties, offering potential therapeutic avenues for managing free radical-induced damage. Future research directions are also discussed to advance our understanding and treatment of free radical-associated diseases.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea
| | - Seon Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Korea
| | - Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Korea
| | - Eun-Kyung Kim
- Nutritional Education Major, Graduate School of Education, Dong-A University, Busan, 49315, Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea.
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea.
| |
Collapse
|
35
|
Zhu Z, Lei M, Guo R, Xu Y, Zhao Y, Wei C, Yang Q, Sun Y. Nicotinamide riboside supplementation ameliorates ovarian dysfunction in a PCOS mouse model. J Ovarian Res 2025; 18:9. [PMID: 39833950 PMCID: PMC11749135 DOI: 10.1186/s13048-025-01596-4] [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: 09/19/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is the leading cause of anovulatory infertility among women of reproductive age, yet the range of effective treatment options remains limited. Our previous study revealed that reduced levels of nicotinamide adenine dinucleotide (NAD+) in ovarian granulosa cells (GCs) of women with PCOS resulted in the accumulation of reactive oxygen species (ROS) and mitochondrial dysfunction. However, it is still uncertain whether increasing NAD+ levels in the ovaries could improve ovarian function in PCOS. In this study, we demonstrated that supplementation with the NAD+ precursor nicotinamide riboside (NR) prevented the decrease in ovarian NAD+ levels, normalized estrous cycle irregularities, and enhanced ovulation potential in dehydroepiandrosterone (DHEA)-induced PCOS mice. Moreover, NR supplementation alleviated ovarian fibrosis and enhanced mitochondrial function in ovarian stromal cells of PCOS mice. Furthermore, NR supplementation improved oocyte quality in PCOS mice, as evidenced by reduced abnormal mitochondrial clustering, enhanced mitochondrial membrane potential, decreased ROS levels, reduced spindle abnormality rates, and increased early embryonic development potential in fertilized oocytes. These findings suggest that supplementing with NAD+ precursors could be a promising therapeutic strategy for addressing ovarian infertility associated with PCOS.
Collapse
Affiliation(s)
- Zhenye Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Lei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruizhi Guo
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yining Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanqing Zhao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenlu Wei
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
36
|
Pellarin I, Dall'Acqua A, Favero A, Segatto I, Rossi V, Crestan N, Karimbayli J, Belletti B, Baldassarre G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct Target Ther 2025; 10:11. [PMID: 39800748 PMCID: PMC11734941 DOI: 10.1038/s41392-024-02080-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive cell division. In reality, the human genome contains 20 different CDKs, which can be divided in at least three different sub-family with different functions, mechanisms of regulation, expression patterns and subcellular localization. Most of these kinases play fundamental roles the normal physiology of eucaryotic cells; therefore, their deregulation is associated with the onset and/or progression of multiple human disease including but not limited to neoplastic and neurodegenerative conditions. Here, we describe the functions of CDKs, categorized into the three main functional groups in which they are classified, highlighting the most relevant pathways that drive their expression and functions. We then discuss the potential roles and deregulation of CDKs in human pathologies, with a particular focus on cancer, the human disease in which CDKs have been most extensively studied and explored as therapeutic targets. Finally, we discuss how CDKs inhibitors have become standard therapies in selected human cancers and propose novel ways of investigation to export their targeting from cancer to other relevant chronic diseases. We hope that the effort we made in collecting all available information on both the prominent and lesser-known CDK family members will help in identify and develop novel areas of research to improve the lives of patients affected by debilitating chronic diseases.
Collapse
Affiliation(s)
- Ilenia Pellarin
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Valentina Rossi
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Nicole Crestan
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Javad Karimbayli
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
37
|
Mohamed HR, Hemdan SHA, El-Sherif AA. Y 2O 3NPs induce selective cytotoxicity, genomic instability, oxidative stress and ROS mediated mitochondrial apoptosis in human epidermoid skin A-431 Cancer cells. Sci Rep 2025; 15:1543. [PMID: 39789066 PMCID: PMC11718274 DOI: 10.1038/s41598-024-82376-w] [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: 09/13/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025] Open
Abstract
Yttrium oxide nanoparticles (Y2O3NPs) have emerged as a promising avenue for cancer therapy, primarily due to their distinctive properties that facilitate selective targeting of cancer cells. Despite their potential, the therapeutic effects of Y2O3NPs on human epidermoid skin cancer remain largely unexplored. This study was thus conducted to investigate the impact of Y2O3NPs on both human skin normal and cancer cells, with an emphasis on assessing their cytotoxicity, genotoxicity, and the mechanisms underlying these effects. Cell viability and apoptosis induction were assessed using the Sulforhodamine B and chromatin diffusion assay, respectively. Reactive oxygen species (ROS) level, mitochondrial membrane potential integrity, oxidative stress markers and expression level of apoptotic and mitochondrial genes were also estimated. Our findings highlight the selective and significant cytotoxicity of Y2O3NPs against human epidermoid A-431 cancer cells. Notably, exposure to five Y2O3NPs concentrations (0.1, 1, 10, 100 and 1000 µg/ml) resulted in a high concentration-dependent reduction in cell viability and a corresponding increase in cell death observed 72 h post-treatment specifically in A-431 cancer cells, while normal skin fibroblast (HSF) cells exhibited minimal toxicity. When A-431 cancer cells were treated with the half-maximal inhibitory concentration (IC50) of Y2O3NPs for 72 h, a significant increase in ROS generation was noted. This led to oxidative stress, along with severe damage to genomic DNA and mitochondrial membrane potential, triggering substantial apoptosis. Furthermore, a concurrent significant upregulation of apoptotic p53 and mitochondrial ND3 genes was observed, coupled with a notable decrease in the anti-apoptotic Bcl2 gene expression.Overall, Y2O3NPs demonstrate considerable promise as a therapeutic agent for skin epidermoid cancer due to their ability to selectively target and induce cytotoxic effects in A-431 cancer cells, all while causing minimal harm to normal HSF cells. This selective cytotoxicity appears to be associated with Y2O3NPs' ability to induce excessive ROS production and subsequent oxidative stress, leading to significant genomic DNA fragmentation, loss of mitochondrial permeability, and alterations in apoptotic and mitochondrial genes' expression, ultimately promoting apoptosis in A-431 cancer cells. These findings establish a foundation for further research into the utilization of Y2O3NPs in targeted cancer therapies and underscore the necessity for ongoing investigation into their safety and efficacy in clinical applications.
Collapse
Affiliation(s)
- Hanan Rh Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Shrouk H A Hemdan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed A El-Sherif
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
38
|
De S, Banerjee S, Rakshit P, Banerjee S, Kumar SKA. Unraveling the Ties: Type 2 Diabetes and Parkinson's Disease - A Nano-Based Targeted Drug Delivery Approach. Curr Diabetes Rev 2025; 21:32-58. [PMID: 38747222 DOI: 10.2174/0115733998291968240429111357] [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: 12/11/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 02/26/2025]
Abstract
The link between Type 2 Diabetes (T2DM) and Parkinson's Disease (PD) dates back to the early 1960s, and ongoing research is exploring this association. PD is linked to dysregulation of dopaminergic pathways, neuroinflammation, decreased PPAR-γ coactivator 1-α, increased phosphoprotein enriched in diabetes, and accelerated α-Syn amyloid fibril production caused by T2DM. This study aims to comprehensively evaluate the T2DM-PD association and risk factors for PD in T2DM individuals. The study reviews existing literature using reputable sources like Scopus, ScienceDirect, and PubMed, revealing a significant association between T2DM and worsened PD symptoms. Genetic profiles of T2DM-PD individuals show similarities, and potential risk factors include insulin-resistance and dysbiosis of the gut-brain microbiome. Anti-diabetic drugs exhibit neuroprotective effects in PD, and nanoscale delivery systems like exosomes, micelles, and liposomes show promise in enhancing drug efficacy by crossing the Blood-Brain Barrier (BBB). Brain targeting for PD uses exosomes, micelles, liposomes, dendrimers, solid lipid nanoparticles, nano-sized polymers, and niosomes to improve medication and gene therapy efficacy. Surface modification of nanocarriers with bioactive compounds (such as angiopep, lactoferrin, and OX26) enhances α-Syn conjugation and BBB permeability. Natural exosomes, though limited, hold potential for investigating DM-PD pathways in clinical research. The study delves into the underlying mechanisms of T2DM and PD and explores current therapeutic approaches in the field of nano-based targeted drug delivery. Emphasis is placed on resolved and ongoing issues in understanding and managing both conditions.
Collapse
Affiliation(s)
- Sourav De
- Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Kolkata, 700126, West Bengal, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - Pallabita Rakshit
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - S K Ashok Kumar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
39
|
Sun Y, Zou Q, Yu H, Yi X, Dou X, Yang Y, Liu Z, Yang H, Jia J, Chen Y, Sun SK, Zhang L. Melanin-like nanoparticles slow cyst growth in ADPKD by dual inhibition of oxidative stress and CREB. EMBO Mol Med 2025; 17:169-192. [PMID: 39567834 PMCID: PMC11730739 DOI: 10.1038/s44321-024-00167-2] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Melanin-like nanoparticles (MNPs) have recently emerged as valuable agents in antioxidant therapy due to their excellent biocompatibility and potent capacity to scavenge various reactive oxygen species (ROS). However, previous studies have mainly focused on acute ROS-related diseases, leaving a knowledge gap regarding their potential in chronic conditions. Furthermore, apart from their well-established antioxidant effects, it remains unclear whether MNPs target other intracellular molecular pathways. In this study, we synthesized ultra-small polyethylene glycol-incorporated Mn2+-chelated MNP (MMPP). We found that MMPP traversed the glomerular filtration barrier and specifically accumulated in renal tubules. Autosomal dominant polycystic kidney disease (ADPKD) is a chronic genetic disorder closely associated with increased oxidative stress and featured by the progressive enlargement of cysts originating from various segments of the renal tubules. Treatment with MMPP markedly attenuated oxidative stress levels, inhibited cyst growth, thereby improving renal function. Interestingly, we found that MMPP effectively inhibits a cyst-promoting gene program downstream of the cAMP-CREB pathway, a crucial signaling pathway implicated in ADPKD progression. Mechanistically, we observed that MMPP directly binds to the bZIP DNA-binding domain of CREB, leading to competitive inhibition of CREB's DNA binding ability and subsequent reduction in CREB target gene expression. In summary, our findings identify an intracellular target of MMPP and demonstrate its potential for treating ADPKD by simultaneously targeting oxidative stress and CREB transcriptional activity.
Collapse
Affiliation(s)
- Yongzhan Sun
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Quan Zou
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huizheng Yu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaoping Yi
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xudan Dou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yu Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiheng Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Hong Yang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junya Jia
- Department of Nephrology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yupeng Chen
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin, China.
| | - Lirong Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
40
|
Agarwal U, Pannu A, Tonk RK. Foreign Contaminants Target Brain Health. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:353-374. [PMID: 39812065 DOI: 10.2174/0118715273338071241213101016] [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: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 01/16/2025]
Abstract
Neurodisease, caused by undesired substances, can lead to mental health conditions like depression, anxiety and neurocognitive problems like dementia. These substances can be referred to as contaminants that can cause damage, corruption, and infection or reduce brain functionality. Contaminants, whether conceptual or physical, have the ability to disrupt many processes. These observations motivate us to investigate contaminants and neurotoxicity collaboratively. This study investigates the link between pollutants and neuro-disease, examining transmission pathways and categorization. It also provides information on resources, causes, and challenges to minimize contamination risks. Contamination may cause various neuro-diseases, including Alzheimer's, Parkinson's, multi-system atrophy, Huntington's, autism spectrum disorder, psychiatric disorder, dementia, meningitis, encephalitis, schizophrenia, anxiety, and depression. The negative effects depend on the nature and extent of exposure. A comprehensive literature search was conducted using databases such as PubMed and Scopus, focusing on studies published till 2024. Studies were selected based on their examination of the relationship between environmental contaminants and brain health, emphasizing transmission pathways and the resulting neurological outcomes. Findings indicate that contaminants can penetrate the blood-brain barrier (BBB) via nasal, gut, and auditory routes, triggering harmful neurophysiological processes. This review highlights the urgent need for increased global awareness, policy interventions, and preventive measures to mitigate the long-term impacts of environmental contaminants on brain health, particularly in emerging nations.
Collapse
Affiliation(s)
- Uma Agarwal
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| | - Arzoo Pannu
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| | - Rajiv Kumar Tonk
- Department of Pharmaceutical Chemistry, Delhi Pharmaceutical Sciences & Research University, Delhi, 110017, India
| |
Collapse
|
41
|
Walker M. Measuring Biomarkers of Oxidative Stress in ME/CFS Patients. Methods Mol Biol 2025; 2920:225-244. [PMID: 40372686 DOI: 10.1007/978-1-0716-4498-0_13] [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: 05/16/2025]
Abstract
Patients with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) have a deficiency in energy production as a result of dysfunctions in their mitochondrial metabolism, defects in the complexes of the electron transport chain, and in the regulation of reactive oxygen species (ROS). This can lead to an imbalance and excess of these species with subsequent modifications of proteins, lipids, and DNA. Oxidative stress is defined as an accumulation of ROS due to a loss of regulation and the subsequent inability to detoxify them. The modifications to the cellular macromolecules by ROS can be used as biomarkers of oxidative stress and so have the potential to monitor the disease course of a condition like ME/CFS. Proteins are especially vulnerable to oxidative stress as amino acid residues are naturally modified as part of cell signaling so, in an imbalance between ROS and antioxidants, proteins become modified at multiple sites potentially altering structure and function. Protein carbonyl modifications are stable and can be measured using 2,4-dinitrophenylhydrazine using a commercial ELISA assay. This has been applied here to immune cell proteins and plasma from ME/CFS patients who had moderate functional activity before and during an exercise protocol, and was shown to have potential as a marker of oxidative stress in these patients. The methods used to measure the DNA modification, 8-hydroxy-2'-deoxyguanosine (8-OHdG) are known to give varied results depending on the technology used. Here, a commercial ELISA assay did not have the sensitivity to detect the modifications in the DNA before and during the exercise protocol of these ME/CFS patients.
Collapse
Affiliation(s)
- Max Walker
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
42
|
Lombardi F, Belmonti S, Sanfilippo A, Borghetti A, Iannone V, Salvo PF, Fabbiani M, Visconti E, Giambenedetto SD. Factors associated with oxidative stress in virologically suppressed people living with HIV on long-term antiretroviral therapy. AIDS Res Ther 2024; 21:100. [PMID: 39734213 DOI: 10.1186/s12981-024-00694-5] [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: 08/28/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Oxidative stress (OS) is the imbalance between oxidant and antioxidant molecules, in favour of oxidants, that has been associated with an increased risk of morbidity and mortality in ART-treated people living with HIV (PLWH). We aimed to assess factors associated with OS in virologically suppressed PLWH on long-term modern ART. METHOD In this cross-sectional study we evaluated OS by measuring both the levels of derivatives-reactive oxygen metabolites (d-ROMs) and the biological antioxidant potential (BAP). We also calculated the BAP/d-ROMs ratio, (OS index, OSi); a cut-off value < 7.3 indicated OS. Factors associated with OS markers were explored by linear regression model. RESULTS We enrolled 299 experienced PLWH with virological suppression (HIV-RNA < 50cps/mL). The mean of the d-ROMs levels was 409 UCARR (95%CI 394-422), whereas the mean of the BAP levels was 1.809 µmol/L (95%CI 1706-1851). The OSi mean value was 4.84, and 91.6% of the participants were below the cut-off value. By regression analysis, higher production of oxidants was associated with female sex (p < 0.001), current exposition to PIs (p = 0.030) and HCV co-infection (p = 0.006). Higher antioxidant capacity was correlated with higher HDL levels (p = 0.001). A lower OSi was associated with female sex (p = 0.003) and the current use of triple vs. dual regimen (p = 0.036). The OSi correlated negatively with cholesterol levels (p = 0.002) and positively with HDL (p < 0.001). CONCLUSIONS Virologically suppressed PLWH on long-term ART showed a marked OS. Female sex, the exposure to PIs, and HCV co-infection were associated with higher oxidants, while higher HDL levels were linked to better antioxidant capacity. Interestingly, dual therapy, especially INSTI-based regimens, was associated with lower oxidative stress compared to triple therapy.
Collapse
Affiliation(s)
- Francesca Lombardi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Malattie Infettive, Largo Agostino Gemelli 8, Rome, 00168, Italia.
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, Roma, Italia.
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy.
| | - Simone Belmonti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Malattie Infettive, Largo Agostino Gemelli 8, Rome, 00168, Italia
| | - Alessia Sanfilippo
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, Roma, Italia
| | | | - Valentina Iannone
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, Roma, Italia
| | | | - Massimiliano Fabbiani
- Università di Pisa, UO Malattie Infettive, AOUP, Pisa, Italy
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Visconti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Malattie Infettive, Largo Agostino Gemelli 8, Rome, 00168, Italia
| | - Simona Di Giambenedetto
- Fondazione Policlinico Universitario A. Gemelli IRCCS, UOC Malattie Infettive, Largo Agostino Gemelli 8, Rome, 00168, Italia
- Dipartimento di Sicurezza e Bioetica, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
43
|
Bogdańska-Chomczyk E, Wojtacha P, Tsai ML, Huang ACW, Kozłowska A. Alterations in Striatal Architecture and Biochemical Markers' Levels During Postnatal Development in the Rat Model of an Attention Deficit/Hyperactivity Disorder (ADHD). Int J Mol Sci 2024; 25:13652. [PMID: 39769412 PMCID: PMC11680085 DOI: 10.3390/ijms252413652] [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: 11/06/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Attention deficit/hyperactivity disorder (ADHD) is defined as a neurodevelopmental condition. The precise underlying mechanisms remain incompletely elucidated. A body of research suggests disruptions in both the cellular architecture and neuronal function within the brain regions of individuals with ADHD, coupled with disturbances in the biochemical parameters. This study seeks to evaluate the morphological characteristics with a volume measurement of the striatal regions and a neuron density assessment within the studied areas across different developmental stages in Spontaneously Hypertensive Rats (SHRs) and Wistar Kyoto Rats (WKYs). Furthermore, the investigation aims to scrutinize the levels and activities of specific markers related to immune function, oxidative stress, and metabolism within the striatum of juvenile and maturing SHRs compared to WKYs. The findings reveal that the most pronounced reductions in striatal volume occur during the juvenile stage in SHRs, alongside alterations in neuronal density within these brain regions compared to WKYs. Additionally, SHRs exhibit heightened levels and activities of various markers, including RAC-alpha serine/threonine-protein kinase (AKT-1), glucocorticoid receptor (GCsRβ), malondialdehyde (MDA), sulfhydryl groups (-SH), glucose (G), iron (Fe), lactate dehydrogenase (LDH). alanine transaminase (ALT), and aspartate transaminase (AST). In summary, notable changes in striatal morphology and elevated levels of inflammatory, oxidative, and metabolic markers within the striatum may be linked to the disrupted brain development and maturation observed in ADHD.
Collapse
Affiliation(s)
- Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| | - Meng-Li Tsai
- Department of Biomechatronic Engineering, National Ilan University, Ylan 26047, Taiwan;
| | | | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Warszawska 30, 10-082 Olsztyn, Poland;
| |
Collapse
|
44
|
Lahlou RA, Gonçalves AC, Bounechada M, Nunes AR, Soeiro P, Alves G, Moreno DA, Garcia-Viguera C, Raposo C, Silvestre S, Rodilla JM, Ismael MI, Silva LR. Antioxidant, Phytochemical, and Pharmacological Properties of Algerian Mentha aquatica Extracts. Antioxidants (Basel) 2024; 13:1512. [PMID: 39765840 PMCID: PMC11673699 DOI: 10.3390/antiox13121512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Water mint (Mentha aquatica) is used in many formulations worldwide as a functional food and natural remedy to treat gastrointestinal disorders, lung diseases, and certain mental disorders such as epilepsy and depression. This study assessed the bioactivity of its infusion extract (INF) and hydroethanolic extract (HE) to highlight its health benefits. These extracts were analyzed for their chemical composition by HPLC-DAD-ESI-MSn, their antioxidant and antidiabetic properties, and their capacities to protect human erythrocytes against induced hemoglobin oxidation and lipid peroxidation. The effect on normal human dermal fibroblast (NHDF) cells and on the N27 rat dopaminergic neuron cell line was also assessed. The chromatographic analysis identified 57 compounds belonging to hydroxycinnamic acids, flavanones, flavone, and isoflavonoids. In respect to the biological potential, the Mentha aquatica extracts revealed a notable capacity for 2,2-diphenyl-1-picrylhydrazyl, nitric oxide, and superoxide radicals, as well as for the inhibition of α-glucosidase action and the protection of human erythrocytes against oxidative damage. Quantification revealed noteworthy phenolic content in both extracts. Additionally, the extracts demonstrated less cytotoxic effects regarding the NHDF and N27 cell lines. Overall, Mentha aquatica presents promising antioxidant activity and a spectrum of potential biological activities, underscoring its significance as a novel antioxidant candidate for applications in animal nutrition, human medicine, and natural product research in the pharmaceutical and nutraceutical industries.
Collapse
Affiliation(s)
- Radhia Aitfella Lahlou
- Chemistry Department, University of Beira Interior, 6201-001 Covilhã, Portugal; (S.S.); (J.M.R.)
- Fiber Materials and Environmental Technologies (FibEnTech), University of Beira Interior, 6201-001 Covilhã, Portugal
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
- SPRINT Sport Physical Activity and Health Research & Innovation Center, Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal
| | - Ana Carolina Gonçalves
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
| | - Mustapha Bounechada
- University Ferhat Abbes Sétif1, Faculty of Natural Sciences and Life, 19000, Algeria;
| | - Ana R. Nunes
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
| | - Pedro Soeiro
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
| | - Gilberto Alves
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
| | - Diego A. Moreno
- Laboratorio de Fitoquímica y Alimentos Saludables” (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (D.A.M.); (C.G.-V.)
| | - Cristina Garcia-Viguera
- Laboratorio de Fitoquímica y Alimentos Saludables” (LabFAS), CSIC, CEBAS, Campus Universitario de Espinardo-25, E-30100 Murcia, Spain; (D.A.M.); (C.G.-V.)
| | - Cesar Raposo
- Mass Spectrometry Service, University of Salamanca, 37007 Salamanca, Spain
| | - Samuel Silvestre
- Chemistry Department, University of Beira Interior, 6201-001 Covilhã, Portugal; (S.S.); (J.M.R.)
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
| | - Jesus M. Rodilla
- Chemistry Department, University of Beira Interior, 6201-001 Covilhã, Portugal; (S.S.); (J.M.R.)
- Fiber Materials and Environmental Technologies (FibEnTech), University of Beira Interior, 6201-001 Covilhã, Portugal
| | - Maria Isabel Ismael
- Chemistry Department, University of Beira Interior, 6201-001 Covilhã, Portugal; (S.S.); (J.M.R.)
- Fiber Materials and Environmental Technologies (FibEnTech), University of Beira Interior, 6201-001 Covilhã, Portugal
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Luís R. Silva
- RISE-Health, Faculdade de Ciências da Saúde, Universidade da Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal; (A.C.G.); (P.S.); (G.A.); (L.R.S.)
- SPRINT Sport Physical Activity and Health Research & Innovation Center, Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal
| |
Collapse
|
45
|
Yahyazadeh A, Gur FM. Promising the potential of β-caryophyllene on mercury chloride-induced alteration in cerebellum and spinal cord of young Wistar albino rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:10175-10189. [PMID: 38995373 PMCID: PMC11582159 DOI: 10.1007/s00210-024-03268-4] [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: 03/28/2024] [Accepted: 06/29/2024] [Indexed: 07/13/2024]
Abstract
Mercury chloride (ME) is a chemical pollutant commonly found in the environment, which can contribute to undesirable health consequence worldwide. The current study investigated the detrimental impact of ME on the cerebellum and spinal cord tissues in 6-8-week-old female rats. We also evaluated the neuroprotective efficacy of β-caryophyllene (BC) against spinal and cerebellar changes caused by ME. Thirty-five young Wistar albino rats were randomly chosen and assigned into five groups: control (CO), olive oil (OI), ME, BC, ME + BC. All samples were analysed by means of unbiased stereological, biochemical, immunohistochemical, and histopathological methods. Our biochemical findings showed that SOD level was significantly increased in the ME group compared to the CO group (p < 0.05). We additionally detected a statistically significant decrease in the number of cerebellar Purkinje cells and granular cells, as well as spinal motor neuron in the ME group compared to the CO group (p < 0.05). In the ME + BC group, the number of Purkinje cells, granular cells, and spinal motor neurons was significantly higher compared to the ME group (p < 0.05). Decreased SOD activity in the ME + BC group was also detected than the ME group (p < 0.05). Immunohistochemical (the tumour necrosis factor-alpha (TNF-α)) and histopathological examinations also exhibited crucial information in each of the group. Taken together, ME exposure was associated with neurotoxicity in the cerebellum and spinal cord tissues. BC treatment also mitigated ME-induced neurological alteration, which may imply its potential therapeutic benefits.
Collapse
Affiliation(s)
- Ahmad Yahyazadeh
- Department of Histology and Embryology, Faculty of Medicine, Karabuk University, Karabuk, Turkey.
| | - Fatih Mehmet Gur
- Department of Histology and Embryology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde, Turkey
| |
Collapse
|
46
|
Chandel J, Naura AS. Dynamics of Inflammatory and Pathological Changes Induced by Single Exposure of Particulate Matter (PM 2.5) in Mice: Potential Implications in COPD. Cell Biochem Biophys 2024; 82:3463-3475. [PMID: 39031246 DOI: 10.1007/s12013-024-01433-3] [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] [Accepted: 07/11/2024] [Indexed: 07/22/2024]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a progressive disorder of lungs marked by chronic bronchitis and emphysema. Particulate matter (PM2.5), a major component of air pollution has been correlated with COPD incidence. The present work aimed to understand dynamics of cellular/molecular players behind PM2.5-mediated COPD pathogenesis in mice by conducting dose and time-course studies. Single intratracheal exposure of PM2.5 at a dose of either 100 or 200 μg induced inflammatory response in lungs at 4 days. Time course studies showed that inflammation once triggered by PM2.5 is progressive in nature as reflected by data on BALF inflammatory cells at 7/14 days. Similarly, various cytokines/chemokines (KC/IL-6/TNF-α/IL-1β/G-CSF/MCP-1) peak at either 7 or 14 days. However, inflammation declined sharply at 21 days. Data on LPO/GSH and activities of SOD/Catalase show induction of continuous oxidative stress in lung tissue. Next, enhanced mtROS in the CD11b+ inflammatory cells confirms the redox imbalance in neutrophils/macrophages. A continuous decline in lung function was observed till 28 days. Further, histological analysis of lung tissues at 28 days confirmed the presence of emphysematous lesions, validating the potency of PM2.5 to cause irreversible damage to lungs through complex interplay of various cellular/molecular players which may be exploited as potential preventive/therapeutic targets.
Collapse
Affiliation(s)
- Jitender Chandel
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Amarjit S Naura
- Department of Biochemistry, Panjab University, Chandigarh, India.
| |
Collapse
|
47
|
Ali M, Tabassum H, Alam MM, Alothaim AS, Al-Malki ES, Jamal A, Parvez S. Valsartan: An Angiotensin Receptor Blocker Modulates BDNF Expression and Provides Neuroprotection Against Cerebral Ischemic Reperfusion Injury. Mol Neurobiol 2024; 61:10805-10819. [PMID: 38789895 DOI: 10.1007/s12035-024-04237-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
AT1 receptor blockers (ARBs) are commonly used drugs to treat cardiovascular disease and hypertension, but research on their impact on brain disorders is unattainable. Valsartan (VAL) is a drug that specifically blocks AT1 receptor. Despite the previous evidence for VAL to provide neuroprotection in case of ischemic reperfusion injury, evaluation of their potential in mitigating mitochondrial dysfunction that causes neuronal cell death and neurobehavioral impairment remains unknown. The aim of this study was to evaluate the therapeutic effect of repurposed drug VAL against ischemic reperfusion injury-induced neuronal alternation. tMCAO surgery was performed to induce focal cerebral ischemic reperfusion injury. Following ischemic reperfusion injury, we analyzed the therapeutic efficacy of VAL by measuring the infarct volume, brain water content, mitochondrial oxidative stress, mitochondrial membrane potential, histopathological architecture, and apoptotic marker protein. Our results showed that VAL administrations (5 and 10 mg/kg b.wt.) mitigated the brain damage, enhanced neurobehavioral outcomes, and alleviated mitochondrial-mediated oxidative damage. In addition to this, our findings demonstrated that VAL administration inhibits neuronal apoptosis by restoring the mitochondrial membrane potential. A follow-up investigation demonstrated that VAL induces BDNF expression and promoted ischemic tolerance via modulating the Akt/p-Creb signaling pathway. In summary, our results suggested that VAL administration provided neuroprotection, ameliorated mitochondrial dysfunction, preserved the integrity of neurons, and lead to functional improvement after ischemic reperfusion injury.
Collapse
Affiliation(s)
- Mubashshir Ali
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Government of India, V. Ramalingaswami Bhawan, New Delhi, 110029, India
| | - Mohammad Mumtaz Alam
- Drug Design and Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Abdulaziz S Alothaim
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia
| | - Esam S Al-Malki
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia
| | - Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Riyadh Region, 11952, Majmaah, Saudi Arabia.
- Health and Basic Science Research Centre, Majmaah University, 11952, Majmaah, Saudi Arabia.
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
48
|
Claro-Cala CM, Rivero-Pino F, Torrecillas-López M, Jimenez-Gonzalez V, Montserrat-de la Paz S. Immunonutrition: future perspective in neurodegenerative disorders. Nutr Neurosci 2024:1-12. [PMID: 39561029 DOI: 10.1080/1028415x.2024.2425565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The relevance of lifestyle, including diet and exercise, has been associated with improved learning and memory capacity, delayed age-related cognitive decline, and a reduced risk of neurodegeneration. Most neurodegenerative diseases are defined as complex multifactorial disorders in which genetic and environmental factors greatly contribute to their onset. Although inflammatory cells produce reactive oxygen species (ROS), oxidative stress itself might exert pro-inflammatory effects and an uncontrolled response could lead to a state of chronic inflammation. Anti-inflammatory dietary approaches unify the disciplines of nutrition, immunity, and neurology. Personalized dietary interventions will be developed based on an individual's genetic makeup, metabolic profile, and gut microbiota composition, thanks to advances in genomics, metabolomics, and microbiome research. The relevance of dietary patterns in decreasing inflammation relies on the role of specific antioxidant nutrients, which might contribute to a decrease in the levels of ROS. This review aims to summarize recent advancements in neuroscience and immunology that have revealed the crucial role that diet and the immune system play in brain function and disease progression. Nutrition influences the immune system, and in turn, the immune system impacts neurological health. This bidirectional relationship suggests that targeted nutritional interventions could modulate immune responses to delay or mitigate the progression of neurodegenerative diseases potentially. This approach focuses on the use of specific nutrients and dietary components that influence the immune system and inflammatory pathway. Key elements of immunonutrition include omega-3 fatty acids, antioxidants, vitamins and various bioactive compounds found in foods.
Collapse
Affiliation(s)
- Carmen María Claro-Cala
- Department of Pharmacology, Pediatrics, and Radiology, School of Medicine, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - Fernando Rivero-Pino
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| | - María Torrecillas-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - Víctor Jimenez-Gonzalez
- CITIUS (Centre for Research, Technology, and Innovation), University of Seville, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
49
|
Barzegar Behrooz A, Aghanoori MR, Nazari M, Latifi-Navid H, Vosoughian F, Anjomani M, Lotfi J, Ahmadiani A, Eliassi A, Nabavizadeh F, Soleimani E, Ghavami S, Khodagholi F, Fahanik-Babaei J. 40 Hz light preserves synaptic plasticity and mitochondrial function in Alzheimer's disease model. Sci Rep 2024; 14:26949. [PMID: 39506052 PMCID: PMC11541745 DOI: 10.1038/s41598-024-78528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Its causes are not fully understood, but it is now known that factors like mitochondrial dysfunction, oxidative stress, and compromised ion channels contribute to its onset and progression. Flickering light therapy has shown promise in AD treatment, though its mechanisms remain unclear. In this study, we used a rat model of streptozotocin (STZ)-induced AD to evaluate the effects of 40 Hz flickering light therapy. Rats received intracerebroventricular (ICV) STZ injections, and 7 days after, they were exposed to 40 Hz flickering light for 15 min daily over seven days. Cognitive and memory functions were assessed using Morris water maze, novel object recognition, and passive avoidance tests. STZ-induced AD rats exhibited cognitive decline, elevated reactive oxygen species, amyloid beta accumulation, decreased serotonin and dopamine levels, and impaired mitochondrial function. However, light therapy prevented these effects, preserving cognitive function and synaptic plasticity. Additionally, flickering light restored mitochondrial metabolites and normalized ATP-insensitive mitochondrial calcium-sensitive potassium (mitoBKCa) channel activity, which was otherwise downregulated in AD rats. Our findings suggest that 40 Hz flickering light therapy could be a promising treatment for neurodegenerative disorders like AD by preserving synaptic and mitochondrial function.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Mohamad-Reza Aghanoori
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary & Alberta Children's Hospital Research Institute, Calgary, AB, T2N 4N1, Canada
| | - Maryam Nazari
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Hamid Latifi-Navid
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Fatemeh Vosoughian
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojdeh Anjomani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jabar Lotfi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Growth and Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Eliassi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nabavizadeh
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Soleimani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, 41-800, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, Canada
- Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Fahanik-Babaei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Baysal M, Karaduman AB, Korkut Çelikateş B, Atlı-Eklioğlu Ö, Ilgın S. Assessment of the toxicity of different antiretroviral drugs and their combinations on Sertoli and Leydig cells. Drug Chem Toxicol 2024; 47:1100-1108. [PMID: 38647040 DOI: 10.1080/01480545.2024.2336506] [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: 01/29/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024]
Abstract
The human immunodeficiency virus continues to pose a significant global public health challenge, affecting millions of individuals. The current treatment strategy has incorporated the utilization of combinations of antiretroviral drugs. The administration of these drugs is associated with many deleterious consequences on several physiological systems, notably the reproductive system. This study aimed to assess the toxic effects of abacavir sulfate, ritonavir, nevirapine, and zidovudine, as well as their combinations, on TM3 Leydig and TM4 Sertoli cells. The cell viability was gauged using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) and neutral red uptake (NRU) assays. Reactive oxygen species (ROS) production was assessed via the 2',7'-dichlorofluorescein diacetate (DCFDA) test, and DNA damage was determined using the comet assay. Results indicated cytotoxic effects at low drug concentrations, both individually and combined. The administration of drugs, individually and in combination, resulted in the production of ROS and caused damage to the DNA at the tested concentrations. In conclusion, the results of this study suggest that the administration of antiretroviral drugs can lead to testicular toxicity by promoting the generation of ROS and DNA damage. Furthermore, it should be noted that the toxicity of antiretroviral drug combinations was shown to be higher compared to that of individual drugs.
Collapse
Affiliation(s)
- Merve Baysal
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Abdullah Burak Karaduman
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Büşra Korkut Çelikateş
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Özlem Atlı-Eklioğlu
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Sinem Ilgın
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|