1
|
Nematalla HA, Elharoun M, Bargash SN, Abd-Alhaseeb MM, Sharafeldin HA, Zewail M, Abbas H, Elsheikh MA. Novel Nose-to-brain delivery of carbenoxolone via mucoadhesive solid lipid nanoparticles for Parkinson's symptoms management: In vitro and in vivo evaluation in a rotenone-induced rat model. Int J Pharm 2025; 670:125197. [PMID: 39793636 DOI: 10.1016/j.ijpharm.2025.125197] [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: 05/05/2024] [Revised: 10/07/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disorder characterized by motor and non-motor symptoms, with limited effective treatment options. This study proposes a novel approach utilizing intranasal delivery of carbenoxolone (CBX) via chitosan-coated solid lipid nanoparticles (CS-coated SLNs) to manage PD symptoms by enhancing CBX delivery and brain targeting. Formulated CS-coated SLNs exhibited favorable quality attributes including particle size (164 ± 0.12 nm), surface charge (18 ± 0.89 mV), high entrapment efficiency (97.98 ± 0.98 %), and sustained drug release profile. In vivo evaluations in a rotenone-induced rat model of PD involved intranasal administration of CBX suspension and CBX-loaded CS-coated SLN (equivalent to 20 mg/kg/day) over four weeks. The CBX nano-formulation group showed significant improvements in motor function, coordination, and balance, as well as modulation of neurotransmitter levels, with increased dopamine and decreased α-synuclein levels compared to the control group. Moreover, the CBX nano-formulation exhibited superior efficacy in reducing neuroinflammation, oxidative stress, and apoptosis markers. Histological examination revealed restored neuronal architecture, suggesting potential neuroprotective effects. In conclusion, mucoadhesive chitosan-coated SLNs offer a promising nasal delivery system overcoming brain drug delivery obstacles facing CBX therapy in PD, paving the way to the development of novel treatments and improved quality of life for PD patients.
Collapse
Affiliation(s)
- Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Mona Elharoun
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Somaia N Bargash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Mohammad M Abd-Alhaseeb
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt; Department of Pharmacology, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Hend A Sharafeldin
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
| | - Mariam Zewail
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Haidy Abbas
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt
| | - Manal A Elsheikh
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour 22511, Egypt.
| |
Collapse
|
2
|
Xu L, Zhang Z, Feng Z, Niu S, Yang L, Xiao B, Jin XM, Ma C, Fan H, Xiao S, Chai Z. Study on the anti-Parkinson's disease activity mechanism and preparation of panaxadiol. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156296. [PMID: 39631295 DOI: 10.1016/j.phymed.2024.156296] [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: 07/05/2024] [Revised: 10/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Traditional Chinese medicine offers unique and valuable resources for the exploration of novel approaches to disease treatment. Panaxadiol, an active compound derived from the transformation of ginsenosides. It has a variety of pharmacological activities. However, there is a lack of research regarding its effects on Parkinson's disease (PD). PURPOSE AND STUDY DESIGN This study investigates the activity and mechanism of panaxadiol in the treatment of PD both in vivo and in vitro. In addition, a new method has been developed to extract panaxadiol. RESULTS The results showed that in vitro, panaxadiol can reduce the viability and cytotoxicity of BV2 cells induced by LPS, and inhibit the production of pro-inflammatory factors through the TLR4/MyD88/NF-κB pathway. In vivo, panaxadiol can improve motor and intestinal dysfunction in PD mice and repair the loss of DA neurons. Further studies have shown that panaxadiol treatment alleviates damage to the blood-brain barrier (BBB), inhibits neuroinflammation in the substantia nigra (SN), and further reduces damage to DA neurons. Subsequently, it was found that panaxadiol alleviated peripheral inflammation and significantly restored the intestinal microbial community. Further mechanistic studies found that panaxadiol treatment inhibited the TLR4/MyD88/NF-κB signaling pathway and its downstream pro-inflammatory products in the SN. In addition, Additionally, the response surface method was employed to identify the optimal conditions for extracting panaxadiol using a deep eutectic solvent (NADES). CONCLUSION In summary, this study has found for the first time that panaxadiol can effectively improve the symptoms of PD by regulating neuroinflammation, repairing the BBB, and gut microbiota. Meanwhile, adopting a green extraction process significantly increases the content of panaxadiol, providing a new direction for the development of PD candidate drugs.
Collapse
Affiliation(s)
- Lei Xu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Zhongjin Zhang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Ziqi Feng
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Sixiang Niu
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Lixia Yang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Baoguo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200025, China
| | - Xiao-Ming Jin
- Department of Anatomy and Cell Biology, Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute. Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cungen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Huijie Fan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Shengnan Xiao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Zhi Chai
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| |
Collapse
|
3
|
Wang Q, Yang S, Zhang X, Zhang S, Chen L, Wang W, Chen N, Yan J. Inflammasomes in neurodegenerative diseases. Transl Neurodegener 2024; 13:65. [PMID: 39710713 PMCID: PMC11665095 DOI: 10.1186/s40035-024-00459-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Inflammasomes represent a crucial component of the innate immune system, which respond to threats by recognizing different molecules. These are known as pathogen-associated molecular patterns (PAMPs) or host-derived damage-associated molecular patterns (DAMPs). In neurodegenerative diseases and neuroinflammation, the accumulation of misfolded proteins, such as beta-amyloid and alpha-synuclein, can lead to inflammasome activation, resulting in the release of interleukin (IL)-1β and IL-18. This activation also induces pyroptosis, the release of inflammatory mediators, and exacerbates neuroinflammation. Increasing evidence suggests that inflammasomes play a pivotal role in neurodegenerative diseases. Therefore, elucidating and investigating the activation and regulation of inflammasomes in these diseases is of paramount importance. This review is primarily focused on evidence indicating that inflammasomes are activated through the canonical pathway in these diseases. Inflammasomes as potential targets for treating neurodegenerative diseases are also discussed.
Collapse
Affiliation(s)
- Qianchen Wang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xuan Zhang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shanshan Zhang
- China Three Gorges University College of Medicine and Health Sciences, Yichang, 443002, China
| | - Liping Chen
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wanxue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jiaqing Yan
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
4
|
Elkady MA, Kabel AM, Dawood LM, Helal AI, Borg HM, Atia HA, Sabry NM, Moustafa NM, Arafa ESA, Alsufyani SE, Arab HH. Targeting the Sirtuin-1/PPAR-Gamma Axis, RAGE/HMGB1/NF-κB Signaling, and the Mitochondrial Functions by Canagliflozin Augments the Protective Effects of Levodopa/Carbidopa in Rotenone-Induced Parkinson's Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1682. [PMID: 39459469 PMCID: PMC11509249 DOI: 10.3390/medicina60101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Parkinson's disease (PD) is a pathological state characterized by a combined set of abnormal movements including slow motion, resting tremors, profound stiffness of skeletal muscles, or obvious abnormalities in posture and gait, together with significant behavioral changes. Until now, no single therapeutic modality was able to provide a complete cure for PD. This work was a trial to assess the immunomodulatory effects of canagliflozin with or without levodopa/carbidopa on rotenone-induced parkinsonism in Balb/c mice. Materials and Methods: In a mouse model of PD, the effect of canagliflozin with or without levodopa/carbidopa was assessed at the behavioral, biochemical, and histopathological levels. Results: The combination of levodopa/carbidopa and canagliflozin significantly mitigated the changes induced by rotenone administration regarding the behavioral tests, striatal dopamine, antioxidant status, Nrf2 content, SIRT-1/PPAR-gamma axis, RAGE/HMGB1/NF-κB signaling, and mitochondrial dysfunction; abrogated the neuroinflammatory responses, and alleviated the histomorphologic changes induced by rotenone administration relative to the groups that received either levodopa/carbidopa or canagliflozin alone. Conclusions: Canagliflozin may represent a new adjuvant therapeutic agent that may add value to the combatting effects of levodopa/carbidopa against the pathological effects of PD.
Collapse
Affiliation(s)
| | - Ahmed M. Kabel
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Lamees M. Dawood
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt;
| | - Azza I. Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Hany M. Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt;
| | - Hanan Abdelmawgoud Atia
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 2440, Saudi Arabia;
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo 35527, Egypt
| | - Nesreen M. Sabry
- Clinical Oncology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Nouran M. Moustafa
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
- Basic Medical Science Department, College of Medicine, Dar Al Uloom University, Riyadh 13314, Saudi Arabia
| | - El-Shaimaa A. Arafa
- College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| |
Collapse
|
5
|
Giradkar V, Mhaske A, Shukla R. Naringenin Nanocrystals Mitigate Rotenone Neurotoxicity in SH-SY5Y Cell Line by Modulating Mitophagy and Oxidative Stress. AAPS PharmSciTech 2024; 25:227. [PMID: 39349907 DOI: 10.1208/s12249-024-02936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Naringenin, a potent antioxidant with anti-apoptotic effects, holds potential in counteracting rotenone-induced neurotoxicity, a model for Parkinson's disease, by reducing oxidative stress and supporting mitochondrial function. Rotenone disrupts ATP production in SH-SY5Y cells through mitochondrial complex-I inhibition, leading to increased reactive oxygen species (ROS) and cellular damage. However, the therapeutic use of naringenin is limited by its poor solubility, low bioavailability, and stability concerns. Nano crystallization of naringenin (NCs), significantly improved its solubility, dissolution rates, and stability for targeted drug delivery. The developed NAR-NC and HSA-NAR-NC formulations exhibit particle sizes of 95.23 nm and 147.89 nm, with zeta potentials of -20.6 mV and -28.5 mV, respectively. These nanocrystals also maintain high drug content and show stability over time, confirming their pharmaceutical viability. In studies using the SH-SY5Y cell line, these modified nanocrystals effectively preserved mitochondrial membrane potential, sustained ATP production, and regulated ROS levels, counteracting the neurotoxic effects of rotenone. Naringenin nanocrystals offer a promising solution for improving the stability and bioavailability of naringenin, with potential therapeutic applications in neurodegenerative diseases.
Collapse
Affiliation(s)
- Vaibhavi Giradkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, 226002, UP, India
| | - Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, 226002, UP, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, 226002, UP, India.
| |
Collapse
|
6
|
Zhao X, Kang Z, Han R, Wang M, Wang Y, Sun X, Wang C, Zhou J, Cao L, Lu M. JWA binding to NCOA4 alleviates degeneration in dopaminergic neurons through suppression of ferritinophagy in Parkinson's disease. Redox Biol 2024; 73:103190. [PMID: 38744191 PMCID: PMC11109895 DOI: 10.1016/j.redox.2024.103190] [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: 04/15/2024] [Revised: 05/11/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024] Open
Abstract
Parkinson's disease (PD) poses a significant challenge in neurodegenerative disorders, characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). The intricate mechanisms orchestrating DA neurodegeneration in PD are not fully understood, necessitating the exploration of innovative therapeutic approaches. Recent studies have implicated ferroptosis as a major contributor to the loss of DA neurons, revealing a complex interplay between iron accumulation and neurodegeneration. However, the sophisticated nature of this process challenges the conventional belief that mere iron removal could effectively prevent DA neuronal ferroptosis. Here, we report JWA, alternatively referred to as ARL6IP5, as a negative regulator of ferroptosis, capable of ameliorating DA neuronal loss in the context of PD. In this study, synchronized expression patterns of JWA and tyrosine hydroxylase (TH) in PD patients and mice were observed, underscoring the importance of JWA for DA neuronal survival. Screening of ferroptosis-related genes unraveled the engagement of iron metabolism in the JWA-dependent inhibition of DA neuronal ferroptosis. Genetic manipulation of JWA provided compelling evidence linking its neuroprotective effects to the attenuation of NCOA4-mediated ferritinophagy. Molecular docking, co-immunoprecipitation, and immunofluorescence studies confirmed that JWA mitigated DA neuronal ferroptosis by occupying the ferritin binding site of NCOA4. Moreover, the JWA-activating compound, JAC4, demonstrated promising neuroprotective effects in cellular and animal PD models by elevating JWA expression, offering a potential avenue for neuroprotection in PD. Collectively, our work establishes JWA as a novel regulator of ferritinophagy, presenting a promising therapeutic target for addressing DA neuronal ferroptosis in PD.
Collapse
Affiliation(s)
- Xinxin Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Zhengwei Kang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Ruixue Han
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Min Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Yueping Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Xin Sun
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Cong Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 211166, Nanjing, China; Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, 213000, Changzhou, China
| | - Lei Cao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China; Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, 213000, Changzhou, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 211166, China; Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, 213000, Changzhou, China.
| |
Collapse
|
7
|
Zhang J, Li X, Zhang Z, Zhang J, Ma L, Wang S, Guo X, Li H, Pan B, Niu Q. Role of the RIP3-PGAM5-Drp1 pathway in aluminum-induced PC12 cells necroptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 270:115860. [PMID: 38142589 DOI: 10.1016/j.ecoenv.2023.115860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 12/10/2023] [Accepted: 12/17/2023] [Indexed: 12/26/2023]
Abstract
Epidemiological studies from diverse global regions suggest a correlation between the accumulation of aluminum in the brain and the onset of various neurodegenerative diseases, including Alzheimer's disease, of which, neuronal cells death happen. Our previous research has found the potential of aluminum to induce neuronal cell death. A comprehensive exploration of the regulatory pathways influenced by aluminum in neuronal cell death could contribute to the development of strategies aimed at preventing the detrimental impact of aluminum on neuronal cells. This study is dedicated to exploring the impact of aluminum on mitochondrial homeostasis through the RIP3-PGAM5-Drp1 pathway, with a specific focus on its potential role in necroptosis. We observed that the inhibition of RIP3 function and the reduction in PGAM5 protein expression both mitigate aluminum-induced necroptosis in PC12 cells and enhance mitochondrial function. However, the inhibition of PGAM5 protein expression does not exert an impact on the expression of RIP3 and MLKL proteins. In summary, our study posits that aluminum can induce necroptosis in PC12 cells through the RIP3-PGAM5-Drp1 pathway.
Collapse
Affiliation(s)
- Jingsi Zhang
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xiaoyan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Zhuoran Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Jintao Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Limin Ma
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Shanshan Wang
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Xin Guo
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi 030001, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Department of Occupational Health, School of Public Health, Jining Medical University, Jining, Shandong 272000, China
| | - Baolong Pan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| |
Collapse
|