1
|
Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ballal S, Sharma R, Debnath S, Sinha A, Rekha A, Khan NH, Alrashoud MM, Kamal M, Imran M. Pathological interplay of NF-κB and M1 macrophages in chronic inflammatory lung diseases. Pathol Res Pract 2025; 269:155903. [PMID: 40081284 DOI: 10.1016/j.prp.2025.155903] [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: 11/29/2024] [Revised: 12/25/2024] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Inflammatory lung diseases such as asthma, chronic obstructive pulmonary disease (COPD), and pulmonary fibrosis depend on the pathology of the nuclear factor kappa B (NF-κB) signalling pathway and M1 macrophage polarization. This review discusses the intimate molecular interactions and processes that modulate NF-κB's promotion of M1 macrophages and chronic inflammation/tissue damage within the confines of this review. NF-κB activation in macrophages produces pro-inflammatory mediators (cytokines - TNFα, IL6, IL1β, and reactive oxygen species (ROS), further increasing airway remodeling and fibrosis. MAPK, JAK-STAT, and PI3K-Akt signalling systems cross-talked with the pathway, amplifying its effect on lung disease progression. Therapeutic strategies focused on inhibiting this axis, including inhibition of NF-κB and small molecule/modulation of macrophage polarization, represent potential ways to attenuate inflammation and promote tissue repair. The potential of precision medicine is illustrated by natural compounds such as curcumin and resveratrol and innovative RNA-based and nanoparticle delivery systems. Despite these challenges, specificity, minimizing systemic side effects, and optimized delivery methods remain difficult. To develop targeted therapies, more research must be conducted to refine targeted approaches, including immune profiling and single-cell analysis. This review aims to advance the management of hard-to-treat inflammatory lung diseases by addressing these complexities.
Collapse
Affiliation(s)
- Fadiyah Jadid Alanazi
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Al Jouf City 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Rajesh Sharma
- Department of Pharmacology, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Sourav Debnath
- Chandigarh pharmacy college, Chandigarh Group of colleges, Jhanjeri, Mohali 140307, Punjab, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - A Rekha
- Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | | | - Muhanad Mubarak Alrashoud
- Department of Inpatient Pharmacy, Dr. Sulaiman Alhabib Hospital, Alhamra Branch, Riyadh 13333, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohd Imran
- Center for Health Research, Northern Border University, Arar, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| |
Collapse
|
2
|
Han HJ, Han J, Choi Y, Hwang GJ, Kim S, Ryoo IJ, Kim BY, Soung NK. A novel tubulin inhibitor, No.07, shows anti-cancer and anti-metastatic effects in colon cancer and tumoroids. Life Sci 2025; 372:123644. [PMID: 40252756 DOI: 10.1016/j.lfs.2025.123644] [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: 01/02/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Colorectal cancer is a highly metastatic disease and the second leading cause of cancer-related death worldwide. Despite the use of various treatment strategies, including chemotherapy and targeted therapy, challenges such as toxicity, drug resistance, and poor response indicate the critical need for new therapeutic agents. Microtubule target agents are one of the major treatment options for chemotherapy in various cancer patients. However, most of these agents are substrates of the MDR1 protein, which leads to the development of multidrug resistance, significantly limiting their effectiveness. Therefore, the development of new drugs is being actively pursued. In this study, we synthesized a novel compound, No.07, which demonstrates significant anti-cancer activity in 3D spheroid models, patient-derived colon cancer organoid models, and mice xenograft models. No.07 directly binds to tubulin dimers, interfering with microtubule polymerization and thereby disrupting tubulin dynamics, ultimately inducing mitotic arrest. Furthermore, No.07 increases mitochondria reactive oxygen species level, leading to the inactivation of the RAF-MEK-ERK signaling cascade, which consequently inhibits metastasis. Notably, Swiss ADME predictions suggest that No.07 is not a substrate of MDR1 and can cross the blood-brain barrier, unlike other microtubule target agents that are limited by MDR1-mediated drug resistance and poor brain penetration. Additionally, experiments using multidrug-resistant cell lines confirmed that No.07 effectively overcomes multidrug resistance, providing a significant improvement over traditionally used chemotherapy agents. In conclusion, No.07 has the potential to address the limitations of existing treatments as a novel therapeutic option.
Collapse
Affiliation(s)
- Ho Jin Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Junyeol Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yerim Choi
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Gwi-Ja Hwang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sumin Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - In-Ja Ryoo
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Bo Yeon Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Nak-Kyun Soung
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
3
|
Maurya R, Sharma A, Naqvi S. Decoding NLRP3 Inflammasome Activation in Alzheimer's Disease: A Focus on Receptor Dynamics. Mol Neurobiol 2025:10.1007/s12035-025-04918-1. [PMID: 40232645 DOI: 10.1007/s12035-025-04918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
Alzheimer's disease (AD) is a leading neurodegenerative disorder marked by progressive cognitive decline and significant neuropsychiatric disturbances. Neuroinflammation, mediated by the NLRP3 inflammasome, is increasingly recognized as a critical factor in AD pathogenesis. The NLRP3 inflammasome, a crucial component of the innate immune system, is activated in response to both pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). In AD, amyloid-beta (Aβ) plaques and tau aggregates act as DAMPs, triggering NLRP3 inflammasome activation in microglia and astrocytes. This activation leads to the production of pro-inflammatory cytokines IL-1β and IL-18, contributing to chronic neuroinflammation and neuronal death. This review explores the intricate mechanisms involved in NLRP3 activation, with a particular focus on TREM-2, Msn Kinase MINK, NF-κB, Toll-like receptors, and P2X7 receptors. Understanding these mechanisms offers insight into the multifaceted regulation of the NLRP3 inflammasome and its impact on AD pathology. By elucidating the roles of TREM-2, MINK1, NF-κB, TLRs, and P2X7 receptors, this review highlights potential therapeutic targets for modulating NLRP3 activity. Targeting these pathways could offer novel strategies for mitigating neuroinflammation and slowing the progression of AD. The interplay between these receptors and signaling pathways underscores the complexity of NLRP3 inflammasome regulation and its significance in AD, providing a foundation for future research aimed at developing effective therapeutic interventions.
Collapse
Affiliation(s)
- Ranika Maurya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Abha Sharma
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India
| | - Saba Naqvi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-R), Lucknow, UP, 226002, India.
| |
Collapse
|
4
|
Saadh MJ, Muhammad FA, Alazzawi TS, Fahdil AA, Athab ZH, Tuxtayev J, Alsaikhan F, Farhood B. Regulation of Apoptotic Pathways by MicroRNAs: A Therapeutic Strategy for Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04833-5. [PMID: 40220245 DOI: 10.1007/s12035-025-04833-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder marked by a gradual decline in memory and cognitive functions. It is characterized by the presence of senile plaques, neurofibrillary tangles, and neuronal degeneration, affecting a significant portion of the human population. A key feature of various nervous system disorders, including AD, is extensive cellular death caused by apoptosis, which affects not only neurons but also glial cells. While apoptosis plays a vital role in eliminating certain cells and supporting normal development, alterations or disruptions in apoptotic pathways can lead to harmful neurodegenerative conditions such as AD. Thus, targeting apoptosis presents a promising therapeutic approach for these diseases. MicroRNAs (miRNAs), a class of non-coding RNA, play diverse roles in cellular functions, including proliferation, gene expression regulation, programmed cell death, intercellular communication, and angiogenesis. By modulating regulatory genes, miRNAs can influence apoptosis, either promoting or inhibiting it. Aberrant expression of miRNAs can impact multiple apoptotic pathways, potentially driving the progression of AD and related health issues. This review summarizes recent research on miRNAs and their dual role in exacerbating or protecting against neural cell damage in AD by altering apoptotic pathways. The regulation of apoptosis by miRNAs offers a prospective therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Jamshid Tuxtayev
- Department of Surgical Diseases, Faculty of Pediatrics, Samarkand State Medical Institute, Samarkand, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
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] [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
|
6
|
Zhang T, Jing J, Liang Y, Luo J, Cheng D, Qin S, Jiang H. Resveratrol-stimulated macrophage exosomes delivering lncRNA Snhg6 inhibit liver fibrosis by modulating the NF-κB pathway. Genomics 2025; 117:111043. [PMID: 40147729 DOI: 10.1016/j.ygeno.2025.111043] [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: 10/16/2024] [Revised: 03/02/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the role of lncRNA Snhg6 in liver fibrosis, delivered by resveratrol-stimulated macrophage exosomes. METHODS Resveratrol-stimulated and unstimulated exosomes were generated from RAW 264.7 cells, confirmed by electron microscopy, nanoparticle analysis, and Western blotting. JS1 cells were used as an HSC model, activated with TGF-β1 and treated with exosomes. Exosome uptake was observed via confocal microscopy, and acta2 expression was measured with immunofluorescence. RNA sequencing and RT-qPCR were used to analyze exosomal lncRNA profiles. KEGG GSEA enrichment was conducted on differentially expressed genes, and nf-κb expression was detected in HSCs using WB. Serum from liver fibrosis patients was analyzed for SNHG6 levels. RESULTS Resveratrol-stimulated exosomes inhibited TGF-β1-induced HSC activation, with 132 differentially expressed lncRNAs, including upregulated Snhg6. NF-κB signaling was downregulated. Silencing Snhg6 weakened this inhibitory effect. CONCLUSION Resveratrol-stimulated macrophage exosomes may inhibit liver fibrosis by delivering lncRNA Snhg6, which suppresses the NF-κB pathway.
Collapse
Affiliation(s)
- Taicheng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jie Jing
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yaodan Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jianming Luo
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China
| | - Dongyu Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China
| | - Shanyu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China.
| | - Haixing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, No. 6, Shuangyong Road, Nanning 530021, Guangxi, China.
| |
Collapse
|
7
|
Pérez R, Burgos V, Cabrera-Pardo JR, Ortiz L, Camins A, Ettcheto M, Schmidt B, Nchiozem-Ngnitedem VA, Paz C. 9-Hydroxyaristoquinolone: A New Indole Alkaloid Isolated from Aristotelia chilensis with Inhibitory Activity of NF-κB in HMC-3 Microglia Cells. Int J Mol Sci 2025; 26:2419. [PMID: 40141065 PMCID: PMC11942453 DOI: 10.3390/ijms26062419] [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: 01/23/2025] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Neurodegenerative diseases are characterized by a progressive process of degeneration and neuronal death in the nervous system, with neuroinflammation being one of the main factors contributing to the progression of these diseases. Aristotelia chilensis (Maqui) is a native tree of Chile used in the Mapuche folk medicine for wounds and digestive treatment. It produces edible black berries with the highest antioxidant capacity among berries, and the Mapuche people used it for producing an alcoholic beverage. The leaves of Maqui contain indole alkaloids with different pharmacological properties that suggest neuroprotective effects. Here, the isolation and chemical characterization of a new alkaloid, named 9-hydroxyaristoquinolone, and the evaluation of its anti-inflammatory activity in the microglial cell line HMC-3, treated with LPS, are reported. 9-Hydroxyaristoquinolone protects microglia from LPS-induced morphological changes at concentrations as low as 1 µM, with a reduction in IKBα-P levels and inhibition of the NF-κB pathway, which was assessed by THP-1 NF-κB dual cell reporter and Western blot in HMC-3 cells. In silico studies suggest that 9-hydroxyaristoquinolone does not induce hepatotoxicity or genotoxicity and exhibits BBB permeability.
Collapse
Affiliation(s)
- Rebeca Pérez
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Viviana Burgos
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Temuco 4780000, Chile;
| | - Jaime R. Cabrera-Pardo
- Laboratorio de Química Aplicada y Sustentable (LabQAS), Departamento de Química, Universidad del Bío-Bío, Avenida Collao 1202, Concepcion 4051381, Chile;
- College of Dental Medicine, Roseman University of Health Sciences, 10894 S. River Front Parkway, South Jordan, UT 84095, USA
| | - Leandro Ortiz
- Instituto de Ciencias Químicas, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile;
| | - Antoni Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.); (M.E.)
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d’Hebron, 171, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, 43204 Reus, Spain
| | - Miren Ettcheto
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Av. de Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.); (M.E.)
- Institut de Neurociències, Universitat de Barcelona (UB), Passeig de la Vall d’Hebron, 171, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CIBERNED), Instituto de Carlos III, Av. Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), Hospital Universitari Sant Joan de Reus, Av. Josep Laporte, 2, 43204 Reus, Spain
| | - Bernd Schmidt
- Institut für Chemie, Universität Potsdam, Karl-Liebknecht-Str. 24-25, D-14476 Potsdam, Germany; (B.S.); (V.-A.N.-N.)
| | | | - Cristian Paz
- Laboratory of Natural Products & Drug Discovery, Center CEBIM, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| |
Collapse
|
8
|
Abdel-Mageed HM. Atopic dermatitis: a comprehensive updated review of this intriguing disease with futuristic insights. Inflammopharmacology 2025; 33:1161-1187. [PMID: 39918744 PMCID: PMC11914373 DOI: 10.1007/s10787-025-01642-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/10/2025] [Indexed: 03/19/2025]
Abstract
Atopic dermatitis (AD) is a paradigmatic prevalent, long-lasting, and inflammatory skin condition with a diverse range of clinical manifestations. The etiology and clinical symptoms of AD are influenced by complex pathophysiological processes, which involve a strong genetic component, epidermal dysfunction, and immunological dysregulation, and a strong influence of other physiological and environmental factors. The FDA has approved targeted and well-tolerated immunomodulators including biologics like dupilumab and crisaborole, and small molecules such as baricitinib, as novel therapies for AD. They effectively treat AD but are too expensive for most patients. The review provides an update on the state of knowledge of AD pathogenesis, discusses the available diagnostic and scoring indices, and provides a scientific foundation for treatment methods for AD. This review also presents data on clinical efficacy of innovative treatments' considering recent guidelines, emphasizing the newest medications and ongoing trials. Finally, the new implication of artificial intelligence (AI) in AD management is explored, where AI can speed up diagnosis and therapy. The PubMed, Google Scholar, and ScienceDirect databases were used for this review.
Collapse
Affiliation(s)
- Heidi M Abdel-Mageed
- Molecular Biology Department, National Research Centre, El Behoth St, Dokki, Giza, Egypt.
| |
Collapse
|
9
|
Mashayekhi-Sardoo H, Hedayati-Moghadam M, Baghcheghi Y. Hippocampal Apoptosis: Molecular Mechanisms Triggered by Toxic Cannabinoid Exposure: A Narrative Review. Neurotoxicology 2025:S0161-813X(25)00019-1. [PMID: 40024347 DOI: 10.1016/j.neuro.2025.02.006] [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: 01/05/2025] [Revised: 02/16/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Hippocampal apoptosis is increasingly recognized as a significant consequence of toxic cannabinoid exposure, with profound implications for cognitive function and mental health. This narrative review comprehensively examines the molecular mechanisms underlying cannabinoid-induced apoptosis, focusing on the interplay of various bioactive compounds and their effects on neuronal integrity. We begin by discussing the key players in cannabinoid biology, followed by a synthesis of findings from animal and clinical studies that highlight the neurotoxic potential of cannabinoids. Central to our analysis are the roles of neuroinflammation and oxidative stress, which exacerbate neuronal damage and contribute to cell death. The activation of cannabinoid receptors, particularly CB1 and CB2, is scrutinized for its dual role in mediating neuroprotective and neurotoxic effects. We explore calcium dysregulation as a critical mechanism that leads to excitotoxicity, mitochondrial dysfunction, and the activation of pro-apoptotic pathways. Additionally, we address the inhibition of anti-apoptotic proteins, induction of endoplasmic reticulum (ER) stress, and disruption of neurotransmitter systems, all of which further facilitate apoptosis in hippocampal neurons. Alterations in neurotrophic factor levels are also examined, as they play a vital role in neuronal survival and plasticity. Ultimately, this review underscores the multifaceted nature of cannabinoid-induced hippocampal apoptosis and calls for further research to elucidate these complex interactions, aiming to inform clinical practices and public health policies regarding cannabinoid use. The findings presented herein highlight the urgent need for a nuanced understanding of the risks associated with cannabinoid exposure, particularly in vulnerable populations.
Collapse
Affiliation(s)
- Habibeh Mashayekhi-Sardoo
- Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran; Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; School of Health, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mahdiyeh Hedayati-Moghadam
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Yousef Baghcheghi
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran; Student Research Committee Jiroft University of Medical Sciences, Jiroft, Iran.
| |
Collapse
|
10
|
Zimmerman AJ, Mangano N, Park G, Kaushal AK, Bergese SD. Glial Modulator Antibiotics for Neuropathic Pain: Current Insights and Future Directions. Pharmaceuticals (Basel) 2025; 18:346. [PMID: 40143124 PMCID: PMC11944926 DOI: 10.3390/ph18030346] [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: 11/16/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Pathological pain is defined as pain that outlives its usefulness as a protective warning system and becomes debilitating, disrupting normal life function. Understanding the mechanism of transition from physiological to pathological pain is essential to provide the effective prevention of chronic pain. The main subcategories of pathological pain are nociceptive pain, neuropathic pain, and nociplastic pain. Glial cells play pivotal roles in the development and maintenance of each of these pathological pain states, specifically neuropathic pain. Consequently, targeting these cells has emerged as a promising therapeutic strategy, as limited efficacy and harmful adverse effects are associated with current pharmacotherapies. This paper aims to review specific antibiotics that modulate glial cells, which can be used to treat neuropathic pain. These antibiotics include minocycline, doxycycline, ceftriaxone, and azithromycin. The potential of these antibiotics appears promising, particularly given the extensive prior research and use of these antibiotics in humans for other illnesses. However, each presents its own set of limitations, ultimately making the translation from preclinical findings to human therapies for neuropathic pain challenging.
Collapse
Affiliation(s)
- Alex J. Zimmerman
- Department of Physical Medicine and Rehabilitation, Stony Brook Medicine, Stony Brook, NY 11794, USA;
| | - Nicholas Mangano
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY 11794, USA;
| | - Grace Park
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Amit K. Kaushal
- Chronic Pain Division, Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY 11794, USA;
| | - Sergio D. Bergese
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY 11794, USA;
| |
Collapse
|
11
|
Kang J, Park C, Yun H, Choi C, Seo W. Engineered Exosomes Carrying Super-Repressor IκB Reduced Biliary Atresia-Induced Liver Fibrosis in Minipig and Mouse Models. Pharmaceutics 2025; 17:264. [PMID: 40006630 PMCID: PMC11859306 DOI: 10.3390/pharmaceutics17020264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background and Aim: Biliary atresia is a rare, progressive disease that affects the bile ducts in newborns. Persistent bile duct obstruction induces various pathological conditions, including jaundice, inflammation, and liver fibrosis; however, the exact pathogenesis of biliary atresia is not yet fully understood. Nuclear factor-κB (NF-κB) is widely acknowledged as a key regulator in the pathogenesis of hepatitis and liver fibrosis, and extensive research has been conducted to develop strategies to effectively inhibit its activity to mitigate liver damage. Exosome-based therapeutic platforms offer targeted NF-κB inhibition with low immunogenicity and enhanced liver-specific delivery. This study aimed to evaluate the therapeutic efficacy of Exo-SrIκB in treating cholestatic liver fibrosis using experimental animal models. Methods: Exo-SrIκB (an exosome-based therapy containing the super-repressor IκB protein) using EXPLOR technology (Exosome engineering for Protein Loading via Optically Reversible protein-protein interactions) to encapsulate the super repressor IκB (SrIκB) within exosomes. The therapeutic efficacy of Exo-SrIκB was assessed in minipig and mouse models with experimentally induced cholestatic liver disease. Results: Administration of Exo-SrIκB significantly attenuated liver fibrosis progression in both animal models by inhibiting NF-κB nuclear translocation and reducing the expression of fibrotic markers. Treated animals exhibited reduced collagen deposition, lower α-SMA levels, and improved hepatic function compared to untreated controls. Conclusion: Exo-SrIκB effectively suppressed NF-κB signaling and alleviated liver fibrosis in experimental cholestatic liver disease models, suggesting that exosome-based therapeutics may offer a targeted and biocompatible application to managing liver fibrosis and other chronic liver diseases.
Collapse
Affiliation(s)
- Jisoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Cheolhyoung Park
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (C.P.); (H.Y.)
| | - Hanoul Yun
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (C.P.); (H.Y.)
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (C.P.); (H.Y.)
| | - Wonhyo Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
- Gradutate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
12
|
Wang YF, Chen CY, Lei L, Zhang Y. Regulation of the microglial polarization for alleviating neuroinflammation in the pathogenesis and therapeutics of major depressive disorder. Life Sci 2025; 362:123373. [PMID: 39756509 DOI: 10.1016/j.lfs.2025.123373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Major depressive disorder (MDD), as a multimodal neuropsychiatric and neurodegenerative illness with high prevalence and disability rates, has become a burden to world health and the economy that affects millions of individuals worldwide. Neuroinflammation, an atypical immune response occurring in the brain, is currently gaining more attention due to its association with MDD. Microglia, as immune sentinels, have a vital function in regulating neuroinflammatory reactions in the immune system of the central nervous system. From the perspective of steady-state branching states, they can transition phenotypes between two extremes, namely, M1 and M2 phenotypes are pro-inflammatory and anti-inflammatory, respectively. It has an intermediate transition state characterized by different transcriptional features and the release of inflammatory mediators. The timing regulation of inflammatory cytokine release is crucial for damage control and guiding microglia back to a steady state. The dysregulation can lead to exorbitant tissue injury and neuronal mortality, and targeting the cellular signaling pathway that serves as the regulatory basis for microglia is considered an essential pathway for treating MDD. However, the specific intervention targets and mechanisms of microglial activation pathways in neuroinflammation are still unclear. Therefore, the present review summarized and discussed various signaling pathways and effective intervention targets that trigger the activation of microglia from its branching state and emphasizes the mechanism of microglia-mediated neuroinflammation associated with MDD.
Collapse
Affiliation(s)
- Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
13
|
Ahn EH, Park JB. Molecular Mechanisms of Alzheimer's Disease Induced by Amyloid-β and Tau Phosphorylation Along with RhoA Activity: Perspective of RhoA/Rho-Associated Protein Kinase Inhibitors for Neuronal Therapy. Cells 2025; 14:89. [PMID: 39851517 PMCID: PMC11764136 DOI: 10.3390/cells14020089] [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: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Amyloid-β peptide (Aβ) is a critical cause of Alzheimer's disease (AD). It is generated from amyloid precursor protein (APP) through cleavages by β-secretase and γ-secretase. γ-Secretase, which includes presenilin, is regulated by several stimuli. Tau protein has also been identified as a significant factor in AD. In particular, Tau phosphorylation is crucial for neuronal impairment, as phosphorylated Tau detaches from microtubules, leading to the formation of neurofibrillary tangles and the destabilization of the microtubule structure. This instability in microtubules damages axons and dendrites, resulting in neuronal impairment. Notably, Aβ is linked to Tau phosphorylation. Another crucial factor in AD is neuroinflammation, primarily occurring in the microglia. Microglia possess several receptors that bind with Aβ, triggering the expression and release of an inflammatory factor, although their main physiological function is to phagocytose debris and pathogens in the brain. NF-κB activation plays a major role in neuroinflammation. Additionally, the production of reactive oxygen species (ROS) in the microglia contributes to this neuroinflammation. In microglia, superoxide is produced through NADPH oxidase, specifically NOX2. Rho GTPases play an essential role in regulating various cellular processes, including cytoskeletal rearrangement, morphology changes, migration, and transcription. The typical function of Rho GTPases involves regulating actin filament formation. Neurons, with their complex processes and synapse connections, rely on cytoskeletal dynamics for structural support. Other brain cells, such as astrocytes, microglia, and oligodendrocytes, also depend on specific cytoskeletal structures to maintain their unique cellular architectures. Thus, the aberrant regulation of Rho GTPases activity can disrupt actin filaments, leading to altered cell morphology, including changes in neuronal processes and synapses, and potentially contributing to brain diseases such as AD.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea;
- Department of Neurology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- ELMED Co., Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| |
Collapse
|
14
|
Zhang XY, Zhang JH, Li XC, Lu H, Liu TCY. Exercise-induced upregulation of TRIM9 attenuates neuroinflammation in Alzheimer's disease-like rat. Int Immunopharmacol 2025; 144:113676. [PMID: 39580859 DOI: 10.1016/j.intimp.2024.113676] [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: 10/04/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVE Exercise exerts protective effects against Alzheimer's disease (AD). However, the factors and mechanisms underlying these effects remain largely unknown. This study aims to elucidate the molecular mechanisms by which exercise exerts its protective effects against AD. METHODS Male 7-week-old Sprague-Dawley rats were randomly allocated to four groups (n = 10 per group): control (CON), exercise control (EXE), sedentary AD model induced by intracerebroventricular streptozotocin (STZ) injection, and AD model with treadmill exercise (EXE + STZ). The exercise groups underwent a 13-week treadmill exercise. An intracerebroventricular injection of STZ was used to induce a rat model of AD. The Barnes maze task was employed as an assessment of spatial learning and memory. Hippocampal tissues from three rats per group was collected for proteomic analysis. Immunofluorescence staining, western blot analysis and polymerase chain reaction were performed for the evaluation of Aβ production, tau hyperphosphorylation, differential protein and corresponding signaling pathway. RESULTS Treadmill exercise could significantly improve STZ-induced cognitive dysfunction and provide neuroprotection by reducing Aβ deposition and tau hyperphosphorylation. Proteomic analysis and further studies demonstrated that treadmill training could significantly increase the expression of tripartite motif-containing 9 (TRIM9). Subsequent research indicated that the upregulation of TRIM9 maybe due, in part,to the inhibition of the NF-κB pathway, thereby reducing the pro-inflammatory factor, and exerting an anti-inflammatory effect. CONCLUSIONS Treadmill exercise attenuates cognitive decline in AD models by upregulating TRIM9 expression, which in turn inhibits NF-κB-mediated neuroinflammation. These findings suggest that TRIM9 may serve as a potential therapeutic target for immunomodulatory strategies against AD.
Collapse
Affiliation(s)
- Xin-Yang Zhang
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Jia-Hao Zhang
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Xiao-Chuan Li
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| | - Hui Lu
- Open Mind Digital Life and Mental Model Laboratory, Shenzhen, Guangzhou 518000, China.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
15
|
Prakash C, Tyagi J, Singh KV, Kumar G, Sharma D. Eugenol attenuates aluminium-induced neurotoxicity in rats by inhibiting the activation of STAT3 and NF-кB. Metab Brain Dis 2025; 40:87. [PMID: 39760810 DOI: 10.1007/s11011-024-01526-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Aluminium is a common metallic toxicant that easily penetrates the brain and exerts severe pathological effects e.g., oxidative stress, inflammation and neurodegeneration. Eugenol is a natural phenolic compound possessing numerous therapeutic properties including antioxidant, anti-inflammatory and neuroprotective. The compound has also been reported to interfere with important transcription factors like STAT3 and NF-кB. Thus, the present study intended to explore the therapeutic potential of eugenol in aluminium neurotoxicity. Rats were administered AlCl3 (100 mg/kg b. wt., orally) and eugenol (200 mg/kg b. wt., orally) alone or in combination for 45 days. The results revealed that AlCl3 administration increases acetylcholinesterase (AChE) activity, lipid peroxidation (LPO), and protein oxidation (PO) along with decreasing superoxide dismutase (SOD) and catalase (CAT) activities, and glutathione (GSH) content in the cortex and hippocampus regions of the brain. Moreover, AlCl3 induces neuronal loss and astroglial activation in both brain areas. The study further revealed that AlCl3 also increases the expression of transcription factors STAT3 and NF-кB in neurons and astrocytes of the cortex and hippocampus. However, co-administration of eugenol with AlCl3 restored the enzymatic activities of AChE, SOD and CAT, and GSH content, and rescued the cortex and hippocampus from LPO, PO, neuronal loss and astroglial activation. Furthermore, the study reported that eugenol reverses the expression pattern of STAT3 and NF-кB in AlCl3-intoxicated rats. In conclusion, the study suggests that eugenol ameliorates oxidative stress, neuronal loss and reactive astrogliosis in aluminium-induced neurotoxicity by inhibiting signalling molecules, STAT3 and NF-кB.
Collapse
Affiliation(s)
- Chandra Prakash
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Jyoti Tyagi
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Kumari Vandana Singh
- Department of Laboratory Medicine, Jaiprakash Narayan Apex Trauma Centre, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Gautam Kumar
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Deepak Sharma
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
16
|
Kannan G, Paul BM, Thangaraj P. Stimulation, regulation, and inflammaging interventions of natural compounds on nuclear factor kappa B (NF-kB) pathway: a comprehensive review. Inflammopharmacology 2025; 33:145-162. [PMID: 39776026 DOI: 10.1007/s10787-024-01635-4] [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: 11/06/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Nuclear factor kappa B (NF-kB) is a kind of transcription factor which resides in cytoplasm of each cell and on activation, it translocates to the nucleus. It is activated by a many inducible agents including endotoxins, inflammatory stimuli, carcinogens, pathogens, nicotine, and tumour promoters, etc. NF-kB is activated by canonical and non-canonical signalling pathways which has different signalling compounds and its biological functions. It controls the expression of 400 different genes including various enzymes, cytokines, viral proteins, regulatory molecules involved in the cell cycle etc. This pathway is linked with various ailments including respiratory diseases, inflammatory diseases, auto immune diseases, cancer and diabetes. NF-kB factor and signalling pathway are the mainstream of the innate and adaptive immune responses. Human subjects have been able to curb inflammation through inflammaging with the help of the phytomolecules interacting with the NF-κB pathway by adjusting the inflammation processes and alleviating aging stresses in cells. They successfully inhibit the activation of NF-κB, thereby curtailing chronic low-grade inflammation underlying both ageing and age-related disease processes. These phytocompounds discussed herewith not only down-regulate NF-κB-dependent pro-inflammatory pathways but also help build resilience at cellular levels, therefore, offering enhanced healthspan with late commencement of inflammaging pathogenesis. This review describes what stimulation and regulation of the Nuclear Factor kappa B (NF-kB) Pathway and its roles in the pathogenesis of human age related diseases. We also review the recent progress in attenuating the molecular mechanisms of the NF-kB Pathway by phytochemicals, which may open up novel therapeutic avenues.
Collapse
Affiliation(s)
- Gowtham Kannan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Benedict Mathews Paul
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India.
| |
Collapse
|
17
|
Asghar MA, Tang S, Wan B, Chen Y, Zhang X, Zhao Q. Valproic acid-induced oxidative stress: Systematic review, meta-analysis and network pharmacology highlights disruption in antioxidant pathways in rodents. Toxicol Appl Pharmacol 2025; 494:117160. [PMID: 39557347 DOI: 10.1016/j.taap.2024.117160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
Valproic acid (VPA) is a widely used antiepileptic drug, but its effects on oxidative stress in rodent models have not been systematically reviewed. This meta-analysis aimed to evaluate the impact of VPA on oxidative stress markers in rodents and explore underlying mechanisms through network pharmacology. A systematic search of PubMed, Web of Science, and PsycINFO (2010-2024) was conducted, following PRISMA and CAMARADES guidelines. Forty-two studies involving 639 rodents were included. Meta-analysis and meta-regression were performed using SPSS and R, and network pharmacology identified key pathways. From 1802 studies, 42 met the criteria, involving 639 rodents. VPA treatment was associated with a significant increase in malondialdehyde (MDA) levels (SMD = 30.45, 95 % CI: 17.64-43.25, P < 0.001) and a decrease in clinically relevant biomarkers, such as superoxide dismutase (SOD) (SMD = -13.22, 95 % CI: -19.39--7.04, P < 0.001), glutathione (GSH) (SMD = -16.97, 95 % CI: -28.13--5.82, P < 0.001), catalase (CAT) (SMD = -9.24, 95 % CI: -13.85--4.62, P < 0.001), glutathione S-transferases (GST) (SMD = -8.82, 95 % CI: -17.40--0.24, P = 0.040), and glutathione peroxidase (GPx) (SMD = -36.05, 95 % CI: -60.72--11.37, P < 0.001). Meta-regression analysis suggested that dosing periods and doses significantly impacted oxidative stress markers. Network pharmacology analysis identified 33 key targets and significant pathways, including MAPK signaling, Toll-like receptor signaling, and TNF signaling. VPA induces oxidative stress in rodent models by increasing MDA and reducing antioxidants, suggesting potential oxidative stress-related side effects in patients.
Collapse
Affiliation(s)
| | - Shixin Tang
- College of Public Health, Chongqing Medical University, Chongqing, PR China
| | - Bing Wan
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Ying Chen
- NMPA Key Laboratory for Quality Control and Evaluation of Pharmaceutical Excipients, Guangdong Institute for Drug Control, Guangzhou 510660, PR China
| | - Xiao Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China
| | - Qinjian Zhao
- College of Pharmacy, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
18
|
Tanaka T, Katayama T, Imai T. Predicting the effects of drugs and unveiling their mechanisms of action using an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG). Comput Biol Med 2025; 184:109419. [PMID: 39556916 DOI: 10.1016/j.compbiomed.2024.109419] [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: 03/11/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Multiple studies have aimed to consolidate drug-related data and predict drug effects. However, most of these studies have focused on integrating diverse data through correlation rather than representing them based on the pharmacodynamic mechanism of action (MOA). It is thus crucial to obtain interpretability to validate prediction results. In this study, we propose a novel framework to construct knowledge graphs that represent pharmacodynamic MOA, predict drug effects, and derive conceivable mechanistic pathways. METHODS AND RESULTS We constructed an interpretable pharmacodynamic mechanism knowledge graph (IPM-KG) by integrating various existing databases and combining them with the approach of this study to automatically fill in the missing data. This yielded a knowledge graph comprising 1455 drugs and 2547 diseases. Additionally, a graph neural network (GNN)-based approach was used to predict therapeutic medication and indication, which outperformed previous approaches that relied on correlation-based knowledge graphs lacking pharmacodynamic MOA representations. Furthermore, we proposed and assessed a method to interpret pharmacodynamic MOA using gene perturbation data. This feasibility study demonstrated the successful derivation of an accurate mechanism in approximately 50 % of cases. Additionally, it facilitated the identification of candidate drugs, which are currently unapproved but exhibit potential for drug repositioning, and their mechanisms of action. CONCLUSIONS This framework not only enables the derivation of highly accurate "drug-indication" predictions but also offers a basic mechanistic understanding, thereby facilitating future drug repositioning efforts.
Collapse
Affiliation(s)
- Tatsuya Tanaka
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Katayama
- Bio Data Science Initiative and Database Center for Life Science, Joint Support-Center for Data Science Research, Research Organization of Information and Systems, Chiba, Japan
| | - Takeshi Imai
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
19
|
Kim ME, Lee JS. Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders. Curr Issues Mol Biol 2024; 47:8. [PMID: 39852123 PMCID: PMC11763386 DOI: 10.3390/cimb47010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. This review explores the cellular and molecular mechanisms underlying neuroinflammation, focusing on the roles of microglia, astrocytes, and peripheral immune cells. Key signaling pathways, including NF-κB, JAK-STAT, and the NLRP3 inflammasome, are discussed alongside emerging regulators such as non-coding RNAs, epigenetic modifications, and the gut-brain axis. The therapeutic landscape is evolving, with traditional anti-inflammatory drugs like NSAIDs and corticosteroids offering limited efficacy in chronic conditions. Immunomodulators, gene and RNA-based therapeutics, and stem cell methods have all shown promise for more specific and effective interventions. Additionally, the modulation of metabolic states and gut microbiota has emerged as a novel strategy to regulate neuroinflammation. Despite significant progress, challenges remain in translating these findings into clinically viable therapies. Future studies should concentrate on integrated, interdisciplinary methods to reduce chronic neuroinflammation and slowing the progression of neurodegenerative disorders, providing opportunities for revolutionary advances in CNS therapies.
Collapse
Affiliation(s)
| | - Jun Sik Lee
- Immunology Research Lab & BK21-Four Educational Research Group for Age-Associated Disorder Control Technology, Department of Biological Science, Chosun University, Gwangju 61452, Republic of Korea;
| |
Collapse
|
20
|
Ghiasi M. Investigating the NF-κB signaling pathway in heart failure: Exploring potential therapeutic approaches. Heliyon 2024; 10:e40812. [PMID: 39717608 PMCID: PMC11664283 DOI: 10.1016/j.heliyon.2024.e40812] [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: 09/17/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Heart failure (HF) syndrome is of great interest as an emerging epidemic. Due to the increasing elderly population worldwide, the total number of HF patients is increasing every day. This disease places a significant economic burden on the healthcare and treatment systems of developing societies, and this situation is very concerning. Despite many advances in the diagnosis and treatment of cardiovascular diseases, HF is still the main cause of death worldwide. This clinical syndrome has many cellular and molecular complications, which are often aggravated by increased levels of pro-inflammatory cytokines, which lead to adverse clinical outcomes. Nuclear factor kappa B (NF-κB), a pivotal family of transcription factors, plays a crucial role in various biological processes, particularly in inflammation, immune response, cell proliferation, and cell survival. Studies show that the NF-κB signaling pathway plays a role in modulating cardiac regeneration, apoptosis, and myocardial fibrosis. It has been found that the NF-κB signaling pathway can affect heart function and HF through the regulation of matrix metalloproteinases and fibrotic mediators. Also, the NF-κB pathway regulates cell activities in cardiac cardiomyocytes and regulates the function of this organ by establishing a precise interaction between apoptosis and pyroptosis. However, the exact molecular mechanisms of this influence have not been well defined and there are many scientific gaps in this matter. This review tries to highlights potential therapeutic strategies to target NF-κB, including the use of anti-inflammatory agents and genetic modulation, which may provide new ways to reduce cardiac fibrosis and improve outcomes in HF patients. Certainly, increasing understanding of the multifaceted role of NF-κB in HF can lead to innovative treatments aimed at reducing the growing number of patients worldwide.
Collapse
Affiliation(s)
- Mohsen Ghiasi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Smail SW. Targeting Neuroinflammation and Apoptosis: Cardamonin's Cognitive Benefits in Alzheimer's 5XFAD Mice. Neurochem Res 2024; 50:57. [PMID: 39673650 DOI: 10.1007/s11064-024-04308-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
This study aimed to evaluate the cognitive-enhancing and neuroprotective effects of cardamonin in the 5XFAD transgenic mouse model of Alzheimer's disease (AD). We treated six-month-old female 5XFAD mice with cardamonin at 5 mg/kg, 10 mg/kg, and 20 mg/kg. Cognitive function was assessed using the Morris Water Maze (MWM) and Novel Object Recognition (NOR) tests. ELISA, western blot, and PCR analyses evaluated amyloid-beta (Aβ) levels, neuroinflammation markers, and apoptosis-related factor expression. All animals survived without toxicity. Cardamonin treatment significantly improved spatial learning and memory retention in MWM and NOR tests, with the 20 mg/kg dose showing the most pronounced effects. Additionally, cardamonin reduced soluble and insoluble Aβ levels in the frontal cortex and hippocampus. The treatment also significantly decreased neuroinflammatory markers, with IL-1β, IL-6, and TNF-α levels dropping substantially at higher doses. Cardamom treatment also normalizes cleaved caspase 3, GFAP, Iba-1, PSD-95, and synaptophysin, which aids in restoring synaptic integrity. Furthermore, cardamonin led to a marked reduction in apoptosis-related gene expression, indicating its potential to mitigate neurodegeneration. Cardamonin demonstrates significant cognitive-enhancing and neuroprotective properties in the 5XFAD mouse model, suggesting its potential as a therapeutic agent for AD. These findings support further investigation into cardamonin's mechanisms and applicability in treating neurodegenerative disorders.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq.
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq.
| |
Collapse
|
22
|
Ntavaroukas P, Michail K, Tsiakalidou R, Stampouloglou E, Tsiggene K, Komiotis D, Georgiou N, Mavromoustakos T, Manta S, Aje D, Michael P, Campbell BJ, Papoutsopoulou S. A Novel Quinoline Inhibitor of the Canonical NF-κB Transcription Factor Pathway. BIOLOGY 2024; 13:910. [PMID: 39596865 PMCID: PMC11591978 DOI: 10.3390/biology13110910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
The NF-κB family of transcription factors is a master regulator of cellular responses during inflammation, and its dysregulation has been linked to chronic inflammatory diseases, such as inflammatory bowel disease. It is therefore of vital importance to design and test new effective NF-κB inhibitors that have the potential to be utilized in clinical practice. In this study, we used a commercial transgenic HeLa cell line as an NF-κB activation reporter to test a novel quinoline molecule, Q3, as a potential inhibitor of the canonical NF-κB pathway. Q3 inhibited NF-κB-induced luciferase in concentrations as low as 5 μM and did not interfere with cell survival or induced cell death. A real-time PCR analysis revealed that Q3 could inhibit the TNF-induced transcription of the luciferase gene, as well as the TNF gene, a known downstream target gene. Immunocytochemistry studies revealed that Q3 moderately interferes with TNF-induced NF-κB nuclear translocation. Moreover, docking and molecular dynamics analyses confirmed that Q3 could potentially modulate transcriptional activity by inhibiting the interaction of NF-κB and DNA. Therefore, Q3 could be potentially developed for further in vivo studies as an NF-κB inhibitor.
Collapse
Affiliation(s)
- Panagiotis Ntavaroukas
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| | - Konstantinos Michail
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece; (N.G.); (T.M.)
| | - Rafaela Tsiakalidou
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| | - Eleni Stampouloglou
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| | - Katerina Tsiggene
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| | - Dimitrios Komiotis
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| | - Nikitas Georgiou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece; (N.G.); (T.M.)
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, 11571 Athens, Greece; (N.G.); (T.M.)
| | - Stella Manta
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
- Laboratory of Organic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Danielle Aje
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK; (D.A.); (P.M.)
| | - Panagiotis Michael
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK; (D.A.); (P.M.)
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Barry J. Campbell
- The Henry Wellcome Laboratories of Molecular & Cellular Gastroenterology, Department of Infection Biology & Microbiomes, Institute of Infection Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 3BX, UK; (D.A.); (P.M.)
| | - Stamatia Papoutsopoulou
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, 41335 Larissa, Greece; (P.N.); (K.M.); (R.T.); (E.S.); (K.T.); (D.K.); (S.M.)
| |
Collapse
|
23
|
Almalki WH, Almujri SS. The impact of NF-κB on inflammatory and angiogenic processes in age-related macular degeneration. Exp Eye Res 2024; 248:110111. [PMID: 39326776 DOI: 10.1016/j.exer.2024.110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Age-related macular degeneration (AMD) is a prominent cause of vision loss, characterized by two different types, dry (atrophic) and wet (neovascular). Dry AMD is distinguished by the progressive deterioration of retinal cells, which ultimately causes a decline in vision. In contrast, wet AMD is defined by the abnormal development of blood vessels underneath the retina, leading to a sudden and severe vision impairment. The course of AMD is primarily driven by chronic inflammation and pathological angiogenesis, in which the NF-κB signaling pathway plays a crucial role. The activation of NF-κB results in the generation of pro-inflammatory cytokines, chemokines, and angiogenic factors like VEGF, which contribute to inflammation and the formation of new blood vessels in AMD. This review analyzes the intricate relationship between NF-κB signaling, inflammation, and angiogenesis in AMD and assesses the possibility of using NF-κB as a target for therapy. The evaluation involves a comprehensive examination of preclinical and clinical evidence that substantiates the effectiveness of NF-κB inhibitors in treating AMD by diminishing inflammation and pathological angiogenesis.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Aseer, Saudi Arabia
| |
Collapse
|
24
|
Kaltschmidt B, Czaniera NJ, Schulten W, Kaltschmidt C. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells. Int J Mol Sci 2024; 25:11353. [PMID: 39518906 PMCID: PMC11545113 DOI: 10.3390/ijms252111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| | - Nele Johanne Czaniera
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| |
Collapse
|
25
|
Bhardwaj S, Grewal AK, Singh S, Dhankar V, Jindal A. An insight into the concept of neuroinflammation and neurodegeneration in Alzheimer's disease: targeting molecular approach Nrf2, NF-κB, and CREB. Inflammopharmacology 2024; 32:2943-2960. [PMID: 38951436 DOI: 10.1007/s10787-024-01502-2] [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: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Alzheimer's disease (AD) is a most prevalent neurologic disorder characterized by cognitive dysfunction, amyloid-β (Aβ) protein accumulation, and excessive neuroinflammation. It affects various life tasks and reduces thinking, memory, capability, reasoning and orientation ability, decision, and language. The major parts responsible for these abnormalities are the cerebral cortex, amygdala, and hippocampus. Excessive inflammatory markers release, and microglial activation affect post-synaptic neurotransmission. Various mechanisms of AD pathogenesis have been explored, but still, there is a need to debate the role of NF-κB, Nrf2, inflammatory markers, CREB signaling, etc. In this review, we have briefly discussed the signaling mechanisms and function of the NF-ĸB signaling pathway, inflammatory mediators, microglia activation, and alteration of autophagy. NF-κB inhibition is a current strategy to counter neuroinflammation and neurodegeneration in the brain of individuals with AD. In clinical trials, numbers of NF-κB modulators are being examined. Recent reports revealed that molecular and cellular pathways initiate complex pathological competencies that cause AD. Moreover, this review will provide extensive knowledge of the cAMP response element binding protein (CREB) and how these nuclear proteins affect neuronal plasticity.
Collapse
Affiliation(s)
- Shaveta Bhardwaj
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Vaibhav Dhankar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Anu Jindal
- G.H.G. Khalsa College of Pharmacy, Gurusar Sudhar, Ludhiana, India
| |
Collapse
|
26
|
Shekho D, Mishra R, Kamal R, Bhatia R, Awasthi A. Breaking Barriers in Alzheimer's Disease: the Role of Advanced Drug Delivery Systems. AAPS PharmSciTech 2024; 25:207. [PMID: 39237748 DOI: 10.1208/s12249-024-02923-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Alzheimer's disease (AD), characterized by cognitive impairment, brain plaques, and tangles, is a global health concern affecting millions. It involves the build-up of amyloid-β (Aβ) and tau proteins, the formation of neuritic plaques and neurofibrillary tangles, cholinergic system dysfunction, genetic variations, and mitochondrial dysfunction. Various signaling pathways and metabolic processes are implicated in AD, along with numerous biomarkers used for diagnosis, risk assessment, and research. Despite these, there is no cure or effective treatment for AD. It is critically important to address this immediately to develop novel drug delivery systems (NDDS) capable of targeting the brain and delivering therapeutic agents to modulate the pathological processes of AD. This review summarizes AD, its pathogenesis, related signaling pathways, biomarkers, conventional treatments, the need for NDDS, and their application in AD treatment. It also covers preclinical, clinical, and ongoing trials, patents, and marketed AD formulations.
Collapse
Affiliation(s)
- Devank Shekho
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Ritika Mishra
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
27
|
Tomar Y, Baidya M, Chadokiya J, Bhatt S, Singhvi G. An overview of Skp2: a promising new therapeutic target of psoriasis. Expert Opin Ther Targets 2024; 28:689-700. [PMID: 39086205 DOI: 10.1080/14728222.2024.2387604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/25/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Psoriasis is a chronic immune-mediated disorder affecting over 2-3% of the population worldwide, significantly impacting quality of life. Despite the availability of various therapeutic interventions, concerns persist regarding lesion recurrence and potential alterations in immune surveillance promoting cancer progression. Recent advancements in understanding cellular and molecular pathways have unveiled key factors in psoriasis etiology, including IL-17, 22, 23, TNF-α, PDE-4, JAK-STAT inhibitors, and AhR agonists. This work explores the potential of S-phase kinase-associated protein 2 (Skp2) as a therapeutic target in psoriasis. AREA COVERED This review covers the current understanding of psoriasis pathophysiology, including immune dysregulation, and the role of keratinocytes and ubiquitin. It also delves into Skp2 role in cell cycle regulation, and its correlation with angiogenesis and ubiquitin in psoriasis. The evolving therapeutic approaches targeting Skp2, including small molecule inhibitors, are also discussed. EXPERT OPINION Targeting Skp2 holds promise for developing novel therapeutic approaches for psoriasis. By modulating Skp2 activity or expression, it may be possible to intervene in inflammatory and proliferative processes underlying the disease. Further research into Skp2 inhibitors and their efficacy in preclinical and clinical settings is warranted to harness the full potential of Skp2 as a therapeutic target in psoriasis management.
Collapse
Affiliation(s)
- Yashika Tomar
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, India
| | - Moushumi Baidya
- Department of Pharmacy, Milestones Institute of Pharmaceutical Sciences, Udaipur, Tripura, India
| | - Jay Chadokiya
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, India
| | - Shvetank Bhatt
- School of Health Sciences and Technology, Dr. Vishwanath Karad MIT World Peace University, Pune, India
| | - Gautam Singhvi
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani, India
| |
Collapse
|
28
|
Wahl D, Risen SJ, Osburn SC, Emge T, Sharma S, Gilberto VS, Chatterjee A, Nagpal P, Moreno JA, LaRocca TJ. Nanoligomers targeting NF-κB and NLRP3 reduce neuroinflammation and improve cognitive function with aging and tauopathy. J Neuroinflammation 2024; 21:182. [PMID: 39068433 PMCID: PMC11283709 DOI: 10.1186/s12974-024-03182-9] [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: 02/13/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Neuroinflammation contributes to impaired cognitive function in brain aging and neurodegenerative disorders like Alzheimer's disease, which is characterized by the aggregation of pathological tau. One major driver of both age- and tau-associated neuroinflammation is the NF-κB and NLRP3 signaling axis. However, current treatments targeting NF-κB or NLRP3 may have adverse/systemic effects, and most have not been clinically translatable. In this study, we tested the efficacy of a novel, nucleic acid therapeutic (Nanoligomer) cocktail specifically targeting both NF-κB and NLRP3 in the brain for reducing neuroinflammation and improving cognitive function in old (aged 19 months) wildtype mice, and in rTg4510 tau pathology mice (aged 2 months). We found that 4 weeks of NF-κB/NLRP3-targeting Nanoligomer treatment strongly reduced neuro-inflammatory cytokine profiles in the brain and improved cognitive-behavioral function in both old and rTg4510 mice. These effects of NF-κB/NLRP3-targeting Nanoligomers were also associated with reduced glial cell activation and pathology, favorable changes in transcriptome signatures of glia-associated inflammation (reduced) and neuronal health (increased), and positive systemic effects. Collectively, our results provide a basis for future translational studies targeting both NF-κB and NLRP3 in the brain, perhaps using Nanoligomers, to inhibit neuroinflammation and improve cognitive function with aging and neurodegeneration.
Collapse
Affiliation(s)
- Devin Wahl
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Sydney J Risen
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Brain Research Center, Colorado State University, Fort Collins, CO, USA
| | - Shelby C Osburn
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Tobias Emge
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA
| | - Sadhana Sharma
- Sachi Bio, Colorado Technology Center, Louisville, CO, USA
| | | | | | | | - Julie A Moreno
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Brain Research Center, Colorado State University, Fort Collins, CO, USA
| | - Thomas J LaRocca
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523, USA.
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
29
|
Zhao T, Zhang X, Cui X, Su S, Li L, Chen Y, Wang N, Sun L, Zhao J, Zhang J, Han X, Cao J. Inhibiting the IRAK4/NF-κB/NLRP3 signaling pathway can reduce pyroptosis in hippocampal neurons and seizure episodes in epilepsy. Exp Neurol 2024; 377:114794. [PMID: 38685307 DOI: 10.1016/j.expneurol.2024.114794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Interleukin-1 receptor-associated kinase 4 (IRAK4) plays an important role in immune modulation in various central nervous system disorders. However, IRAK4 has not been reported in epilepsy models in animal and clinical studies, nor has its involvement in regulating pyroptosis in epilepsy. METHOD First, we performed transcriptome sequencing, quantitative real-time polymerase chain reaction, and western blot analysis on the hippocampal tissues of refractory epilepsy patients to measure the mRNA and protein levels of IRAK4 and pyroptosis-related proteins. Second, we successfully established a pentylenetetrazol (PTZ)-induced seizure mouse model. We conducted behavioral tests, electroencephalography, virus injection, and molecular biology experiments to investigate the role of IRAK4 in seizure activity regulation. RESULTS IRAK4 is upregulated in the hippocampus of epilepsy patients and PTZ-induced seizure model mice. IRAK4 expression is observed in the hilar neurons of PTZ-induced mice. Knocking down IRAK4 in PTZ-induced mice downregulated pyroptosis-related protein expression and alleviated seizure activity. Overexpressing IRAK4 in naive mice upregulated pyroptosis-related protein expression and increased PTZ-induced abnormal neuronal discharges. IRAK4 and NF-κB were found to bind to each other in patient hippocampal tissue samples. Pyrrolidine dithiocarbamate reversed the pyroptosis-related protein expression increase caused by PTZ. PF-06650833 alleviated seizure activity and inhibited pyroptosis in PTZ-induced seizure mice. CONCLUSION IRAK4 plays a key role in the pathological process of epilepsy, and its potential mechanism may be related to pyroptosis mediated by the NF-κB/NLRP3 signaling pathway. PF-06650833 has potential as a therapeutic agent for alleviating epilepsy.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xuefei Zhang
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaoxiao Cui
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Songxue Su
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Lei Li
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Yanan Chen
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Na Wang
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Lei Sun
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Jiewen Zhang
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Xiong Han
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| | - Jing Cao
- Department of Neurology and Basic Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China.
| |
Collapse
|
30
|
Wahl D, Risen SJ, Osburn SC, Emge T, Sharma S, Gilberto VS, Chatterjee A, Nagpal P, Moreno JA, LaRocca TJ. Nanoligomers targeting NF-κB and NLRP3 reduce neuroinflammation and improve cognitive function with aging and tauopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.03.578493. [PMID: 38370618 PMCID: PMC10871285 DOI: 10.1101/2024.02.03.578493] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Neuroinflammation contributes to impaired cognitive function in brain aging and neurodegenerative disorders like Alzheimer's disease, which is characterized by the aggregation of pathological tau. One major driver of both age- and tau-associated neuroinflammation is the NF-κB and NLRP3 signaling axis. However, current treatments targeting NF-κB or NLRP3 may have adverse/systemic effects, and most have not been clinically translatable. In this study, we tested the efficacy of a novel, nucleic acid therapeutic (Nanoligomer) cocktail specifically targeting both NF-κB and NLRP3 in the brain for reducing neuroinflammation and improving cognitive function in old (aged 19 months) wildtype mice, and in rTg4510 tau pathology mice (aged 2 months). We found that 4 weeks of NF-κB/NLRP3-targeting Nanoligomer treatment strongly reduced neuro-inflammatory cytokine profiles in the brain and improved cognitive-behavioral function in both old and rTg4510 mice. These effects of NF-κB/NLRP3-targeting Nanoligomers were also associated with reduced glial cell activation and pathology, favorable changes in transcriptome signatures of glia-associated inflammation (reduced) and neuronal health (increased), and positive systemic effects. Collectively, our results provide a basis for future translational studies targeting both NF-κB and NLRP3 in the brain, perhaps using Nanoligomers, to inhibit neuroinflammation and improve cognitive function with aging and neurodegeneration.
Collapse
|
31
|
Góral I, Wichur T, Sługocka E, Godyń J, Szałaj N, Zaręba P, Głuch-Lutwin M, Mordyl B, Panek D, Więckowska A. Connecting GSK-3β Inhibitory Activity with IKK-β or ROCK-1 Inhibition to Target Tau Aggregation and Neuroinflammation in Alzheimer's Disease-Discovery, In Vitro and In Cellulo Activity of Thiazole-Based Inhibitors. Molecules 2024; 29:2616. [PMID: 38893493 PMCID: PMC11173485 DOI: 10.3390/molecules29112616] [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: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
GSK-3β, IKK-β, and ROCK-1 kinases are implicated in the pathomechanism of Alzheimer's disease due to their involvement in the misfolding and accumulation of amyloid β (Aβ) and tau proteins, as well as inflammatory processes. Among these kinases, GSK-3β plays the most crucial role. In this study, we present compound 62, a novel, remarkably potent, competitive GSK-3β inhibitor (IC50 = 8 nM, Ki = 2 nM) that also exhibits additional ROCK-1 inhibitory activity (IC50 = 2.3 µM) and demonstrates anti-inflammatory and neuroprotective properties. Compound 62 effectively suppresses the production of nitric oxide (NO) and pro-inflammatory cytokines in the lipopolysaccharide-induced model of inflammation in the microglial BV-2 cell line. Furthermore, it shows neuroprotective effects in an okadaic-acid-induced tau hyperphosphorylation cell model of neurodegeneration. The compound also demonstrates the potential for further development, characterized by its chemical and metabolic stability in mouse microsomes and fair solubility.
Collapse
Affiliation(s)
- Izabella Góral
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Tomasz Wichur
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Emilia Sługocka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| |
Collapse
|
32
|
Dai H, Jiang Y, Liu S, Li D, Zhang X. Dietary flavonoids modulate the gut microbiota: A new perspective on improving autism spectrum disorder through the gut-brain axis. Food Res Int 2024; 186:114404. [PMID: 38729686 DOI: 10.1016/j.foodres.2024.114404] [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: 12/19/2023] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.
Collapse
Affiliation(s)
- Haochen Dai
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Yuhan Jiang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China
| | - Shuxun Liu
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, PR China.
| | - Dandan Li
- Sinograin Chengdu Storage Research Institute Co., Ltd, Chengdu 610091, PR China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
33
|
Li MC, Li MZ, Lin ZY, Zhuang YM, Wang HY, Jia JT, Lu Y, Wang ZJ, Zou HY, Zhao H. Buyang Huanwu Decoction promotes neurovascular remodeling by modulating astrocyte and microglia polarization in ischemic stroke rats. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117620. [PMID: 38141792 DOI: 10.1016/j.jep.2023.117620] [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: 10/26/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Buyang Huanwu Decoction (BYHWD), one of the most commonly utilized traditional Chinese medicine prescription for treatment of cerebral ischemic stroke. However, the understanding of BYHWD on neurovascular repair following cerebral ischemia is so far limited. AIM OF THE STUDY This research investigated the influence of BYHWD on neurovascular remodeling by magnetic resonance imaging (MRI) technology and revealed the potential neurovascular repair mechanism underlying post-treatment with BYHWD after ischemic stroke. MATERIALS AND METHODS Male Sprague-Dawley rats were utilized as an ischemic stroke model by permanent occlusion of the middle cerebral artery (MCAO). BYHWD was intragastrically administrated once daily for 30 days straight. Multimodal MRI was performed to detect brain tissue injuries, axonal microstructural damages, cerebral blood flow and intracranial vessels on the 30th day after BYHWD treatment. Proangiogenic factors, axonal/synaptic plasticity-related factors, energy transporters and adenosine monophosphate-activated protein kinase (AMPK) signal pathway were evaluated using western blot. Double immunofluorescent staining and western blot were applied to evaluate astrocytes and microglia polarization. RESULTS Administration of BYHWD significantly alleviated infarct volume and brain tissue injuries and ameliorated microstructural damages, accompanied with improved axonal/synaptic plasticity-related factors, axonal growth guidance factors and decreased axonal growth inhibitors. Meanwhile, BYHWD remarkably improved cerebral blood flow, cerebral vascular signal and promoted the expression of proangiogenic factors. Particularly, treatment with BYHWD obviously suppressed astrocytes A1 and microglia M1 polarization accompanied with promoted astrocyte A2 and microglia M2 polarization. Furthermore, BYHWD effectively improved energy transporters. Especially, BYHWD markedly increased expression of phosphorylated AMPK, cyclic AMP-response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF) accompanied by inactivation of the NF-κB. CONCLUSION Taken together, these findings identified that the beneficial roles of BYHWD on neurovascular remodeling were related to AMPK pathways -mediated energy transporters and NFκB/CREB pathways.
Collapse
Affiliation(s)
- Ming-Cong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Man-Zhong Li
- Department of pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China; Beijing Key Laboratory of Bio-characteristic Profiling for Evaluation of Rational Drug Use, Beijing, 100038, China
| | - Zi-Yue Lin
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yu-Ming Zhuang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Han-Yu Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Jing-Ting Jia
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Zhan-Jing Wang
- Medical Imaging laboratory of Core Facility Center, Capital Medical University, Beijing, 100069, China
| | - Hai-Yan Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| |
Collapse
|
34
|
Al-Kuraishy HM, Jabir MS, Albuhadily AK, Al-Gareeb AI, Jawad SF, Swelum AA, Hadi NR. Role of ketogenic diet in neurodegenerative diseases focusing on Alzheimer diseases: The guardian angle. Ageing Res Rev 2024; 95:102233. [PMID: 38360180 DOI: 10.1016/j.arr.2024.102233] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
The ketogenic diet (KD) is a low-carbohydrate, adequate protein and high-fat diet. KD is primarily used to treat refractory epilepsy. KD was shown to be effective in treating different neurodegenerative diseases. Alzheimer disease (AD) is the first common neurodegenerative disease in the world characterized by memory and cognitive impairment. However, the underlying mechanism of KD in controlling of AD and other neurodegenerative diseases are not discussed widely. Therefore, this review aims to revise the fundamental mechanism of KD in different neurodegenerative diseases focusing on the AD. KD induces a fasting-like which modulates the central and peripheral metabolism by regulating mitochondrial dysfunction, oxidative stress, inflammation, gut-flora, and autophagy in different neurodegenerative diseases. Different studies highlighted that KD improves AD neuropathology by regulating synaptic neurotransmission and inhibiting of neuroinflammation and oxidative stress. In conclusion, KD improves cognitive function and attenuates the progression of AD neuropathology by reducing oxidative stress, mitochondrial dysfunction, and enhancing neuronal autophagy and brain BDNF.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Majid S Jabir
- Department of Applied Science, University of Technology Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq; Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-iraq, PO.Box13, Kufa, Iraq.
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, 51001, Iraq.
| | - Ayman A Swelum
- Department of Animal Production, King Saud University, Riyadh, Saudi Arabia.
| | | |
Collapse
|
35
|
Dama A, Shpati K, Daliu P, Dumur S, Gorica E, Santini A. Targeting Metabolic Diseases: The Role of Nutraceuticals in Modulating Oxidative Stress and Inflammation. Nutrients 2024; 16:507. [PMID: 38398830 PMCID: PMC10891887 DOI: 10.3390/nu16040507] [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: 01/14/2024] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
The escalating prevalence of metabolic and cardiometabolic disorders, often characterized by oxidative stress and chronic inflammation, poses significant health challenges globally. As the traditional therapeutic approaches may sometimes fall short in managing these health conditions, attention is growing toward nutraceuticals worldwide; with compounds being obtained from natural sources with potential therapeutic beneficial effects being shown to potentially support and, in some cases, replace pharmacological treatments, especially for individuals who do not qualify for conventional pharmacological treatments. This review delves into the burgeoning field of nutraceutical-based pharmacological modulation as a promising strategy for attenuating oxidative stress and inflammation in metabolic and cardiometabolic disorders. Drawing from an extensive body of research, the review showcases various nutraceutical agents, such as polyphenols, omega-3 fatty acids, and antioxidants, which exhibit antioxidative and anti-inflammatory properties. All these can be classified as novel nutraceutical-based drugs that are capable of regulating pathways to mitigate oxidative-stress- and inflammation-associated metabolic diseases. By exploring the mechanisms through which nutraceuticals interact with oxidative stress pathways and immune responses, this review highlights their potential to restore redox balance and temper chronic inflammation. Additionally, the challenges and prospects of nutraceutical-based interventions are discussed, encompassing bioavailability enhancement, personalized treatment approaches, and clinical translation. Through a comprehensive analysis of the latest scientific reports, this article underscores the potential of nutraceutical-based pharmacological treatment modulation as a novel avenue to fight oxidative stress and inflammation in the complex landscape of metabolic disorders, particularly accentuating their impact on cardiovascular health.
Collapse
Affiliation(s)
- Aida Dama
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, 1017 Tirana, Albania; (A.D.); (K.S.); (P.D.)
| | - Kleva Shpati
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, 1017 Tirana, Albania; (A.D.); (K.S.); (P.D.)
| | - Patricia Daliu
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, 1017 Tirana, Albania; (A.D.); (K.S.); (P.D.)
| | - Seyma Dumur
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, 34408 Istanbul, Türkiye;
| | - Era Gorica
- Department of Pharmacy, Faculty of Medical Sciences, Albanian University, 1017 Tirana, Albania; (A.D.); (K.S.); (P.D.)
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, 8952 Zurich, Switzerland
| | - Antonello Santini
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
| |
Collapse
|
36
|
Alharbi KM, Alshehri SA, Almarwani WA, Aljohani KK, Albalawi AZ, Alatawi AS, Al-Atwi SM, Alhwyty LS, Hassan HM, Al-Gayyar MMH. Effects of Cycloastragenol on Alzheimer's Disease in Rats by Reducing Oxidative Stress, Inflammation, and Apoptosis. Curr Alzheimer Res 2024; 21:141-154. [PMID: 38766828 DOI: 10.2174/0115672050315334240508162754] [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: 03/08/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND As individuals age, they may develop Alzheimer's disease (AD), which is characterized by difficulties in speech, memory loss, and other issues related to neural function. Cycloastragenol is an active ingredient of Astragalus trojanus and has been used to treat inflammation, aging, heart disease, and cancer. OBJECTIVES This study aimed to explore the potential therapeutic benefits of cycloastragenol in rats with experimentally induced AD. Moreover, the underlying molecular mechanisms were also evaluated by measuring Nrf2 and HO-1, which are involved in oxidative stress, NFκB and TNF-α, which are involved in inflammation, and BCL2, BAX, and caspase-3, which are involved in apoptosis. METHODS Sprague-Dawley rats were given 70 mg/kg of aluminum chloride intraperitoneally daily for six weeks to induce AD. Following AD induction, the rats were given 25 mg/kg of cycloastragenol daily by oral gavage for three weeks. Hippocampal sections were stained with hematoxylin/ eosin and with anti-caspase-3 antibodies. The Nrf2, HO-1, NFκB, TNF-α, BCL2, BAX, and caspase-3 gene expressions and protein levels in the samples were analyzed. RESULTS Cycloastragenol significantly improved rats' behavioral test performance. It also strengthened the organization of the hippocampus. Cycloastragenol significantly improved behavioral performance and improved hippocampal structure in rats. It caused a marked decrease in the expression of NFκB, TNF-α, BAX, and caspase-3, which was associated with an increase in the expression of BCL2, Nrf2, and HO-1. CONCLUSION Cycloastragenol improved the structure of the hippocampus in rats with AD. It enhanced the outcomes of behavioral tests, decreased the concentration of AChE in the brain, and exerted antioxidant and anti-inflammatory effects. Antiapoptotic effects were also noted, leading to significant improvements in cognitive function, memory, and behavior in treated rats.
Collapse
Affiliation(s)
- Kadi M Alharbi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Shahad A Alshehri
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Wasayf A Almarwani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Khulud K Aljohani
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Ajwan Z Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Areej S Alatawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Shekha M Al-Atwi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Lama S Alhwyty
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Hanan M Hassan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa City, Egypt
| | - Mohammed M H Al-Gayyar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia
| |
Collapse
|
37
|
Balistreri CR, Monastero R. Neuroinflammation and Neurodegenerative Diseases: How Much Do We Still Not Know? Brain Sci 2023; 14:19. [PMID: 38248234 PMCID: PMC10812964 DOI: 10.3390/brainsci14010019] [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: 11/27/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
The term "neuroinflammation" defines the typical inflammatory response of the brain closely related to the onset of many neurodegenerative diseases (NDs). Neuroinflammation is well known, but its mechanisms and pathways are not entirely comprehended. Some progresses have been achieved through many efforts and research. Consequently, new cellular and molecular mechanisms, diverse and conventional, are emerging. In listing some of those that will be the subject of our description and discussion, essential are the important roles of peripheral and infiltrated monocytes and clonotypic cells, alterations in the gut-brain axis, dysregulation of the apelinergic system, alterations in the endothelial glycocalyx of the endothelial component of neuronal vascular units, variations in expression of some genes and levels of the encoding molecules by the action of microRNAs (miRNAs), or other epigenetic factors and distinctive transcriptional factors, as well as the role of autophagy, ferroptosis, sex differences, and modifications in the circadian cycle. Such mechanisms can add significantly to understanding the complex etiological puzzle of neuroinflammation and ND. In addition, they could represent biomarkers and targets of ND, which is increasing in the elderly.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy
| | - Roberto Monastero
- Unit of Neurology & Neuro-Physiopathology, Department of Biomedicine, Neuroscience, and Advanced Diagnostics (Bi.N.D), University of Palermo, Via La Loggia 1, 90129 Palermo, Italy;
| |
Collapse
|
38
|
Suresh S, Singh S A, Rushendran R, Vellapandian C, Prajapati B. Alzheimer's disease: the role of extrinsic factors in its development, an investigation of the environmental enigma. Front Neurol 2023; 14:1303111. [PMID: 38125832 PMCID: PMC10730937 DOI: 10.3389/fneur.2023.1303111] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
In the realm of Alzheimer's disease, the most prevalent form of dementia, the impact of environmental factors has ignited intense curiosity due to its substantial burden on global health. Recent investigations have unveiled these environmental factors as key contributors, shedding new light on their profound influence. Notably, emerging evidence highlights the detrimental role of various environmental contaminants in the incidence and progression of Alzheimer's disease. These contaminants encompass a broad spectrum, including air pollutants laden with ozone, neurotoxic metals like lead, aluminum, manganese, and cadmium, pesticides with their insidious effects, and the ubiquitous presence of plastics and microplastics. By meticulously delving into the intricate web connecting environmental pollutants and this devastating neurological disorder, this comprehensive chapter takes a deep dive into their involvement as significant risk factors for Alzheimer's disease. Furthermore, it explores the underlying molecular mechanisms through which these contaminants exert their influence, aiming to unravel the complex interactions that drive the pathogenesis of the disease. Additionally, this chapter proposes potential strategies to mitigate the detrimental effects of these environmental contaminants on brain health, with the ultimate goal of restoring and preserving typical cognitive function. Through this comprehensive exploration, we aim to enhance our understanding of the multifaceted relationship between neurotoxins and Alzheimer's disease, providing a solid foundation for developing innovative in-vivo models and advancing our knowledge of the intricate pathological processes underlying this debilitating condition.
Collapse
Affiliation(s)
- Swathi Suresh
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ankul Singh S
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Rapuru Rushendran
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Chitra Vellapandian
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Bhupendra Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat, India
| |
Collapse
|