1
|
Jin Y, Ji W, Zhang L, Dang D, Yu B, Zhang X, Zhang Y, Li J, Zhang Y, Yang R, Yang H, Chen S, Wang F, Duan G. Arginine depletion-induced autophagy and metabolic dysregulation are involved in the disease severity of hand, foot, and mouth disease. Virulence 2025; 16:2440541. [PMID: 39731500 DOI: 10.1080/21505594.2024.2440541] [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: 06/04/2024] [Revised: 10/22/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024] Open
Abstract
Amino acid metabolism provides significant insight into the development and prevention of many viral diseases. Therefore, the present study aimed to compare the amino acid profiles of hand, foot, and mouth disease (HFMD) patients with those of healthy individuals and to further reveal the molecular mechanisms of HFMD severity. Using UPLC-MS/MS, we determined the plasma amino acid expression profiles of pediatric patients with HFMD (mild, n = 42; severe, n = 43) and healthy controls (n = 25). Brain tissues from CVA6-infected mice were examined using untargeted metabolomics. Several amino acids were significantly different between the three groups. Pathway analysis revealed that arginine, proline, and tryptophan metabolism are implicated in the pathogenesis of HFMD. A similar arginine depletion was observed in the brain tissues of CVA6-infected mice. Importantly, L-arginine supplementation improved the survival rate of CVA6-infected mice, inhibited virus multiplication, and reduced pathological autophagy associated with mTOR-autophagy pathway in the brain. Collectively, arginine, as the hub amino acid metabolite of the mammalian target of rapamycin (mTOR) signaling pathway affecting autophagy, plays an important role in the pathogenesis of severe HFMD. L-arginine supplementation may serve as a potential therapeutic option for critical patients with HFMD.
Collapse
Affiliation(s)
- Yuefei Jin
- Department of Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Wangquan Ji
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Dejian Dang
- Department of Infection Control, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bingqing Yu
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaolong Zhang
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Yuxiang Zhang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaqi Li
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaodong Zhang
- Henan International Joint Laboratory of Children's Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Rongxin Yang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Yang
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Shuaiyin Chen
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Fang Wang
- Department of Infectious Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Guangcai Duan
- Department of Epidemiology, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
2
|
Ntetsika T, Catrina SB, Markaki I. Understanding the link between type 2 diabetes mellitus and Parkinson's disease: role of brain insulin resistance. Neural Regen Res 2025; 20:3113-3123. [PMID: 39715083 PMCID: PMC11881720 DOI: 10.4103/nrr.nrr-d-23-01910] [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: 11/21/2023] [Revised: 01/22/2024] [Accepted: 03/03/2024] [Indexed: 12/25/2024] Open
Abstract
Type 2 diabetes mellitus and Parkinson's disease are chronic diseases linked to a growing pandemic that affects older adults and causes significant socio-economic burden. Epidemiological data supporting a close relationship between these two aging-related diseases have resulted in the investigation of shared pathophysiological molecular mechanisms. Impaired insulin signaling in the brain has gained increasing attention during the last decade and has been suggested to contribute to the development of Parkinson's disease through the dysregulation of several pathological processes. The contribution of type 2 diabetes mellitus and insulin resistance in neurodegeneration in Parkinson's disease, with emphasis on brain insulin resistance, is extensively discussed in this article and new therapeutic strategies targeting this pathological link are presented and reviewed.
Collapse
Affiliation(s)
- Theodora Ntetsika
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Diabetes, Academic Specialist Center, Stockholm, Sweden
| | - Ioanna Markaki
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Center for Neurology, Academic Specialist Center, Stockholm, Sweden
| |
Collapse
|
3
|
Sisto A, van Wermeskerken T, Pancher M, Gatto P, Asselbergh B, Assunção Carreira ÁS, De Winter V, Adami V, Provenzani A, Timmerman V. Autophagy induction by piplartine ameliorates axonal degeneration caused by mutant HSPB1 and HSPB8 in Charcot-Marie-Tooth type 2 neuropathies. Autophagy 2025; 21:1116-1143. [PMID: 39698979 PMCID: PMC12013449 DOI: 10.1080/15548627.2024.2439649] [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: 01/18/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
HSPB1 [heat shock protein family B (small) member 1] and HSPB8 are essential molecular chaperones for neuronal proteostasis, as they prevent protein aggregation. Mutant HSPB1 and HSPB8 primarily harm peripheral neurons, resulting in axonal Charcot-Marie-Tooth neuropathies (CMT2). Macroautophagy/autophagy is a shared mechanism by which HSPB1 and HSPB8 mutations cause neuronal dysfunction. Autophagosome formation is reduced in mutant HSPB1-induced pluripotent stem-cell-derived motor neurons from CMT type 2F patients. Likewise, the HSPB8K141N knockin mouse model, mimicking CMT type 2 L, exhibits axonal degeneration and muscle atrophy, with SQSTM1/p62-positive deposits. We show here that mouse embryonic fibroblasts isolated from a HSPB8K141N/green fluorescent protein (GFP)-LC3 model have diminished autophagosome production under conditions of MTOR inhibition. To correct the autophagic deficits in the HSPB1 and HSPB8 models, we screened by high-throughput autophagosome quantification the repurposing Spectrum Collection library for molecules that could boost the autophagic activity above the canonical MTOR inhibition. Hit compounds were validated on motor neurons obtained by differentiation of HSPB1P182L and HSPB8K141N patient-derived induced pluripotent stem cells, focusing on autophagy induction as well as neurite network density, axonal degeneration, and mitochondrial morphology. We identified molecules that specifically stimulate autophagosome formation in the HSPB8K141N cells, without affecting autophagy flux. Two top lead compounds induced autophagy and reduced axonal degeneration, thus promoting neuronal network maturation in the CMT2 patient-derived motor neurons. Based on these findings, the phenotypical screen revealed that piplartine rescued autophagy deficiencies in both the HSPB1 and HSPB8 models, demonstrating autophagy induction as an effective therapeutic strategy for CMT neuropathies and other chaperonopathies.
Collapse
Affiliation(s)
- Angela Sisto
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Tamira van Wermeskerken
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Pamela Gatto
- HTS Core Facility, University of Trento, Trento, Italy
| | - Bob Asselbergh
- Neuromics Support Facility, VIB - Center for Molecular Neurology, Antwerp, Belgium
- Neuromics Support Facility, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Vicky De Winter
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | | | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of Neuromuscular Pathology, Institute Born Bunge, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
4
|
Utpal BK, Mokhfi FZ, Zehravi M, Sweilam SH, Gupta JK, Kareemulla S, C RD, Rao AA, Kumar VV, Krosuri P, Prasad D, Khan SL, Roy SC, Rab SO, Alshehri MA, Emran TB. Resveratrol: A Natural Compound Targeting the PI3K/Akt/mTOR Pathway in Neurological Diseases. Mol Neurobiol 2025; 62:5579-5608. [PMID: 39578340 DOI: 10.1007/s12035-024-04608-4] [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: 03/21/2024] [Accepted: 11/01/2024] [Indexed: 11/24/2024]
Abstract
Neurological diseases (NDs), including neurodegenerative disorders and acute injuries, are a significant global health concern. The PI3K/Akt/mTOR pathway, a crucial signaling cascade, is responsible for the survival of cells, proliferation, and metabolism. Dysregulation of this pathway has been linked to neurological conditions, indicating its potential as a vital target for therapeutic approaches. Resveratrol (RSV), a natural compound found in berries, peanuts, and red grapes, has antioxidant, anti-cancer, and anti-inflammatory effects. Its ability to modulate the PI3K/Akt/mTOR pathway has been interesting in NDs. Studies have shown that RSV can activate the PI3K/Akt pathway, promoting cell survival and inhibiting apoptosis of neuronal cells. Its impact on mTOR, a downstream effector of Akt, further contributes to its neuroprotective effects. RSV's ability to restore autophagic flux presents a promising avenue for therapeutic intervention. Its anti-inflammatory properties suppress inflammatory responses by inhibiting key signaling molecules within the pathway. Additionally, RSV's role in enhancing mitochondrial function contributes to its neuroprotective profile. This study highlights RSV's potential as a multifaceted therapeutic agent in NDs, specifically by PI3K/Akt/mTOR pathway modulation. Additional investigation is required to optimize its therapeutic capacity in diverse neurological conditions.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Fatima Zohra Mokhfi
- Laboratory of AgroBiotechnology and Nutrition in Semi Arid Zones, Faculty of Nature and Life Sciences, University of Ibn Khaldoun, Tiaret, Algeria
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia.
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| | - Jeetendra Kumar Gupta
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University Mathura, Chaumuha, Mathura, Uttar Pradesh, 281406, India
| | - Shaik Kareemulla
- Department of Pharmacy Practice, Malla Reddy College of Pharmacy (MRCP), Kompally, Secunderabad, Telangana, 500100, India
| | - Ronald Darwin C
- Department of Pharmacology, School of Pharmaceutical Sciences, Technology and Advanced Studies (VISTAS), Vels Institute of Science, Pallavaram, Chennai, 600117, India
| | - A Anka Rao
- KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Guntur, Andhra Pradesh, 522502, India
| | - Voleti Vijaya Kumar
- Department of Pharmaceutics, School of Pharmacy, Satyabhama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Pavankumar Krosuri
- Department of Pharmaceutics, Santhiram College of Pharmacy, NH40, Nandyal, Andhra Pradesh, 518112, India
| | - Dharani Prasad
- Depertment of Pharmacology Mohan Babu University MB School of Pharmaceutical Sciences, Erstwhile Sree Vidyaniketan College of Pharmacy, Tirupati, Andhra Pradesh, 517102, India
| | - Sharukh L Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa, 413520, Maharashtra, India
| | - Sajib Chandra Roy
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Ali Alshehri
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, 1207, Bangladesh.
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh.
| |
Collapse
|
5
|
Mao S, Yu N, Wang W, Mao Y, Du Y, Zhao Q, Gu X, Kang J. Ubiquitin-specific peptidase 10 attenuates bleomycin-induced pulmonary fibrosis via modulating autophagy depending on sirtuin 6-mediated AKT/mTOR. Cell Biol Toxicol 2025; 41:73. [PMID: 40278953 DOI: 10.1007/s10565-025-10031-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF), characterized by fibroblast activation and collagen deposition, is a progressive lung disease that lacks effective interventions. Ubiquitin-specific peptidase 10 (USP10) acts as a multifunctional player in inflammatory response and progression of cancers, the effect on pulmonary fibrosis is unknown. Here, we demonstrated downregulated expression of USP10 in fibrotic lung tissues of IPF patients. In the current study, lung tissues were collected at the end of weeks 1, 2, or 3 post bleomycin (BLM)-intratracheal delivery. Consistently, USP10 expression levels were reduced after BLM challenge in a time-dependent manner. Mice treated with lentivirus overexpressing USP10 exhibited mitigative lung injury and reduced collagen deposition. USP10 overexpression enhanced autophagy in BLM-treated mouse lungs. Interestingly, the protective effect of USP10 was attenuated as the pulmonary autophagy flux was blocked by autophagy inhibitor 3-methyladenine (3-MA). Primary human and mouse lung fibroblasts were treated with pro-fibrotic TGF-β1 to verify the role of USP10 in vitro. Mechanically, the deubiquitinating enzyme USP10 interacted with Sirtuin 6 (Sirt6) and inhibited its degradation. Furthermore, USP10 overexpression inhibited the activation of Sirt6-mediated AKT/mTOR pathway in both lung tissues and fibroblasts. Our findings suggest that USP10 might attenuate pulmonary fibrosis through the promotion of Sirt6/AKT/mTOR-mediated autophagy. These data prioritize USP10 as a therapeutic target for treating IPF.
Collapse
Affiliation(s)
- Shitao Mao
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Yu
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yikai Mao
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Du
- Department of Otolaryngology Head and Neck Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qihe Zhao
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiu Gu
- Department of Pulmonary and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jian Kang
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Rossato RC, Salles GR, Albuquerque AL, Porcionatto MA, Granato AEC, Ulrich H, Dos Santos MIB, Pacheco-Soares C. Photobiomodulation by LED 660 nm and Taurine against H 2O 2 oxidative stress in SH-SY5Y cells. Lasers Med Sci 2025; 40:211. [PMID: 40274660 DOI: 10.1007/s10103-025-04467-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
Alzheimer's Disease (AD) is a progressive uncurable neurodegenerative pathology affecting millions worldwide. Photobiomodulation and Taurine are promising alternatives for preventing and reducing the rapid progression of neurodegeneration, stimulating the reconstructing of neural tissue structures, especially improving mitochondrial activity, which is highly impaired in AD. In this study, the mitochondrial effects of Taurine combined with light emitting diode (LED) irradiation were evaluated on human neuroblastoma cells (SH-SY5Y), under oxidative stress condition by hydrogen peroxide (H2O2) exposure, a considerable modulator in AD. We evaluated LED irradiation at the wavelength of 660 nm and Taurine under different concentrations before and together with exposing SH-SY5Y cells to different concentrations of H2O2, assessing mitochondrial activity by the MTT colorimetric test and labeling live cells mitochondria by the fluorescent probe MitoTracker. Cell viability was also evaluated by the trypan blue exclusion assay, and cellular morphological structures were imaged by scanning electron microscopy (SEM). Neuroprotective effects were achieved by both LED irradiation and LED irradiation + Taurine when cells were exposed to them before H2O2-induced stress. Comparing both agents, LED irradiation at 660 nm is sufficient to improve mitochondrial activity, however, healthy mitochondrial morphology was only observed when cells were treated with Taurine together with LED irradiation, representing affordable candidates that act in synergy against oxidative stress, one of the main contributors to neurodegeneration.
Collapse
Affiliation(s)
- Rafaella Carvalho Rossato
- Universidade Do Vale Do Paraíba, Av. Shishima Hifumi, 2911, Urbanova, São José Dos Campos, SP, 12244‑000, Brazil
| | - Geisa Rodrigues Salles
- Universidade Do Vale Do Paraíba, Av. Shishima Hifumi, 2911, Urbanova, São José Dos Campos, SP, 12244‑000, Brazil.
- Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Pedro de Toledo, 669, Vila Clementino, São Paulo, SP, 04039 - 032, Brazil.
| | - Amanda Lira Albuquerque
- Universidade Do Vale Do Paraíba, Av. Shishima Hifumi, 2911, Urbanova, São José Dos Campos, SP, 12244‑000, Brazil
| | - Marimélia Aparecida Porcionatto
- Escola Paulista de Medicina, Universidade Federal de São Paulo, R. Pedro de Toledo, 669, Vila Clementino, São Paulo, SP, 04039 - 032, Brazil
- National Institute of Science and Technology in Modeling Human Complex Diseases With 3D Platforms (INCT Model 3D), São Paulo, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes, 748, São Paulo, SP, 05508 - 000, Brazil
| | | | - Cristina Pacheco-Soares
- Universidade Do Vale Do Paraíba, Av. Shishima Hifumi, 2911, Urbanova, São José Dos Campos, SP, 12244‑000, Brazil.
| |
Collapse
|
7
|
Wang L, Guo H, Zhao W, Wang J, Cao X. Oxiracetam ameliorates neurological function after traumatic brain injury through competing endogenous RNA regulatory network. Psychopharmacology (Berl) 2025:10.1007/s00213-025-06797-9. [PMID: 40272502 DOI: 10.1007/s00213-025-06797-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
RATIONALE Oxiracetam (ORC) has been demonstrated to improve neurological function resulting from traumatic brain injury (TBI). OBJECTIVES This study aims to explore the precise molecular mechanism of ORC in the treatment of TBI. METHODS TBI rat model was established and treated with ORC. Modified Garcia score, rotarod test and HE staining were employed to evaluate the neuroprotective effects of ORC. Subsequently, RNA-seq was conducted on the hippocampus of sham, TBI and ORC rats to identify differential expression (DE) lncRNAs and mRNAs. Functional analysis of DE lncRNAs and mRNAs was performed. The real-time quantitative polymerase chain reaction (qRT-PCR) was used to determine the expression of DE lncRNAs and DE mRNAs. Western blot was performed to explore important pathway in ceRNA networks. RESULTS ORC has been demonstrated to effectively improve neurological function in TBI rats. A total of 10 ORC-treated DE lncRNAs and 61 DE mRNAs were obtained. A co-expression network comprising 79 lncRNA-mRNA pairs associated with the treatment of ORC was constructed. Furthermore, an lncRNA-miRNA-mRNA regulated ceRNA network was constructed, comprising 15 mRNAs, 41 miRNAs and 10 lncRNAs. Functional enrichment, qRT-PCR, and Western blot analysis showed that ORC improve neurological function of TBI rats by regulating multiple signaling pathways, including the JAK-STAT/PI3K-Akt pathway, as well as affecting the expression of key genes Prlr, Cdkn1a, and Cldn1. CONCLUSION Our study reveals the mechanism of ORC therapy in TBI rats, which mainly relies on the regulation of the JAK-STAT/PI3K-Akt pathway and the influence on the expression of key genes Prlr, Cdkn1a, and Cldn1.
Collapse
Affiliation(s)
- Liyi Wang
- Hospital Infection-Control Department, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Han Guo
- Department of Oral Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Weidong Zhao
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Jiahao Wang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, 050000, China
| | - Xuhua Cao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, Hebei Province, 050000, China.
| |
Collapse
|
8
|
Oh SW, Yu E, Park SH, Kwon K, Lee JH, Ha H, Kim G, Shin HS, Min S, Song M, Cho JY, Lee J. Ammonium chloride, an environmental pollutant, disrupts melanocyte biology through the regulation of melanosome and autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118214. [PMID: 40262244 DOI: 10.1016/j.ecoenv.2025.118214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Ammonium chloride is an indoor environmental pollutant released due to industrial emissions, concrete, indoor bacteria, or other sources. In this study, we characterized molecular mechanisms of ammonium chloride-induced cell damage in melanocyte cells, which are a critical effector for pigmentation. Specifically, we investigated the effects of ammonium chloride on pigmentation and its underlying mechanisms, including its involvement in melanogenesis and autophagy. Based on the experiments, we elucidated that ammonium chloride induced and increased melanogenesis by upregulating MITF via AKT-mediated melanogenic signaling pathways. Moreover, ammonium chloride did not exhibit lysosomotropic activity and inhibited autophagy by activating the AKT-mTOR signaling pathway, suggesting that the pigment-regulating mechanism of ammonium chloride was associated with autophagy in pigmented cells. The findings of this study offer new perspectives on the mechanisms involved in ammonium chloride-induced pigmentation and propose a potential approach to mitigate ammonium chloride-induced side effects like hyperpigmentation and hyperammonemia by employing a combined autophagy inducer.
Collapse
Affiliation(s)
- Sae Woong Oh
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Eunbi Yu
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong City 30016, South Korea
| | - Kitae Kwon
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Jung Hyun Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Heejun Ha
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Gyeonghyeon Kim
- Department of MetaBioHealth, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Hee Seon Shin
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Seokhyeon Min
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea
| | - Minkyung Song
- Integrative Research of T cells Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Department of Biopharmaceutical Convergence, Sungkyunkwan University, Gyunggi Do 16419, South Korea.
| | - Jae Youl Cho
- Molecular Immunology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea.
| | - Jongsung Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon City, Gyunggi Do 16419, South Korea.
| |
Collapse
|
9
|
Zhu L, Yang Y, Tan J, Lin Y, Qing J, Li X, Zeng L. Effect of 2,5-hexanedione on rat ovarian granulosa cell apoptosis involves endoplasmic reticulum stress-dependent m-TOR signaling pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:319-328. [PMID: 39668517 DOI: 10.1080/15287394.2024.2438832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Occupational exposure to N-hexane/2,5-hexanedione (2,5-HD) was found to adversely affect reproductive functions in females. However, there are few studies regarding the mechanisms underlying reproductive system damage initiated by 2,5-HD. Several studies demonstrated that 2,5-HD exerts hormonal dysfunctions in females by promoting apoptosis using rat ovarian granulosa cells (GCs) as a model. The endoplasmic reticulum (ER) plays a key role in cellular processes such as protein folding and modification, Ca2+ storage, and lipid synthesis, which are known to involve the activation of stress (ERS)-dependent m-TOR signaling pathway. Thus, the aim of this study was to examine the effects of 2,5-HD on ER and the associated activation of stress (ERS)-dependent m-TOR signaling pathway resulting in consequent apoptosis of ovarian GCs. Data demonstrated that after intraperitoneal treatment with 100, 200, or 400 mg/kg 2,5-HD for 6 consecutive weeks, 5 times per week, a decrease in body weight, ovarian weight, and relative ovary weight was found. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay showed that 2,5-HD promoted apoptosis of ovarian GCs, which involved enhanced relative protein expression levels of m-TOR/p-mTOR. Our findings demonstrated that 2,5-HD (1) elevated expression levels of pro-apoptosis-related genes Bax and Caspase 3, (2) decreased expression levels of the anti-apoptosis gene Bcl-2, and (3) activated the protein expression of glucose-regulatory protein 78 (GRP78), inositol-requiring enzyme-1 (IRE1), and c-Jun terminal kinase (JNK) associated with increased apoptosis. Evidence indicates that chronic exposure to 2,5-HD induced apoptosis of ovarian GCs, and the possible mechanism underlying this effect involves the ERS-dependent m-TOR signaling pathway.
Collapse
Affiliation(s)
- Lemei Zhu
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Yue Yang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Jingsi Tan
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Yibo Lin
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Jiaqi Qing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Xin Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Lingfeng Zeng
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
- Department of Pharmacology and Toxicology, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| |
Collapse
|
10
|
Mussalo L, Afonin AM, Zavodna T, Krejcik Z, Honkova K, Fayad C, Shahbaz MA, Kalapudas J, Penttilä E, Löppönen H, Koivisto AM, Malm T, Topinka J, Jalava P, Lampinen R, Kanninen KM. Traffic-related ultrafine particles influence gene regulation in olfactory mucosa cells altering PI3K/AKT signaling. ENVIRONMENT INTERNATIONAL 2025; 199:109484. [PMID: 40273555 DOI: 10.1016/j.envint.2025.109484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/10/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Traffic-related ultrafine particles (UFPs) are an emerging health concern affecting the brain and increasing the risk of Alzheimer's disease (AD). PI3K/AKT signaling is known to contribute to neuronal survival and to be altered in AD. The nasal olfactory mucosa (OM) is a sensory tissue exposed directly to ambient air, and a starting point for olfactory neural circuits towards the brain. Evidence of air pollution-induced transcriptional regulation via microRNAs (miRNA) and DNA methylation (DNAmet) is accumulating and air pollutant-mediated disturbances in PI3K/AKT signaling have been reported. By utilizing a highly translational human-based in vitro model of OM, we aimed to investigate possible gene regulatory mechanisms in PI3K/AKT signaling induced by UFPs, and to compare the responses between cognitively healthy and individuals with AD. miRNA expression was analyzed using next-generation sequencing (NGS) and chip-based methylation analysis was performed to detect differentially methylated loci (DML). These data were combined with previously published transcriptomics analysis (mRNA) to construct an mRNA-miRNA-DNAmet-integrative network. Protein level changes were studied by immunoassays. We observed UFP-induced reductions in viability and increases in oxidative stress and DNA damage without eminent cell death. Integrative network analysis revealed multiple connections of miRNAs to differentially expressed genes in the PI3K/AKT pathway, and effects were most prominent in AD cells. Similarly, in AD cells DML were identified in transcription factor and apoptosis genes, downstream of PI3K/AKT signaling. Conclusively, traffic-related UFPs influence gene regulation of PI3K/AKT signaling to modulate OM cell survival, with existing AD pathology resulting in heightened vulnerability to UFP effects.
Collapse
Affiliation(s)
- Laura Mussalo
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Alexey M Afonin
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Tana Zavodna
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Zdenek Krejcik
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Katerina Honkova
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Claire Fayad
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Muhammad Ali Shahbaz
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Juho Kalapudas
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Elina Penttilä
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Heikki Löppönen
- Department of Otorhinolaryngology, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anne M Koivisto
- Department of Neurology, Neuro Centre, Kuopio University Hospital, 70210 Kuopio, Finland; Brain Research Unit, Department of Neurology, School of Medicine, University of Eastern Finland, 70210 Kuopio, Finland; Department of Neurology and Geriatrics, Helsinki University Hospital and Neurosciences, Faculty of Medicine, University of Helsinki 00014 Helsinki, Finland
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Pasi Jalava
- Inhalation Toxicology Laboratory, Department of Environmental and Biological Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Riikka Lampinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland
| | - Katja M Kanninen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland.
| |
Collapse
|
11
|
Cao J, Qin X, Yang H, Liu C, Cheng T. Dimm targets GDAP2 to regulate larval development in the silkworm, Bombyx mori. INSECT SCIENCE 2025. [PMID: 40205793 DOI: 10.1111/1744-7917.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/16/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
The basic helix-loop-helix (bHLH) domain transcription factors precisely regulate various developmental processes in insects. Dimm, a specific bHLH transcription factor, integrates the insulin/insulin-like growth factor signaling (IIS) and juvenile hormone signaling (JHS) pathways to modulate larval development in silkworms. However, the molecular mechanisms underlying this regulation are not yet fully understood. This study aimed to determine the targets of Dimm through which it regulates larval development. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) revealed ganglioside-induced differentiation-associated protein 2 (GDAP2) as a direct downstream target gene of Dimm. Further study showed that Dimm directly binds to an enhancer element located in the second intron of the GDAP2 gene to promote its transcription. GDAP2 exhibited widespread expression across different stages and tissues of silkworms, regulated by both the IIS and the JHS pathways. The systemic knockout of GDAP2 leads to delayed larval development with a significant reduction in body weight; moreover, larval development was arrested at the 4th-instar stage. Further investigation unveiled that the inhibition of the ecdysone and innate immune signaling pathways in the mutant line led to abnormal larval development. A systematic investigation of the biological functions of GDAP2 offers valuable insights into the mechanism by which Dimm integrates IIS and JHS pathways to regulate the larval development of silkworms.
Collapse
Affiliation(s)
- Jun Cao
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Xiaodan Qin
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Hongguo Yang
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Chun Liu
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| | - Tingcai Cheng
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
| |
Collapse
|
12
|
Huang L, Li Q, Wu J, He Y, Huang J, Xie S, Yang C, Ruan Q, Zhou Z, Deng M. Galangin reduces MPTP-induced dopamine neuron injury via the autophagy dependent-PI3K/AKT pathway. Front Aging Neurosci 2025; 17:1568002. [PMID: 40256391 PMCID: PMC12006098 DOI: 10.3389/fnagi.2025.1568002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Research has confirmed that Galangin can attenuate autophagy and protect dopaminergic neurons. This study aims to clarify whether Galangin attenuates dopaminergic neuron injury by regulating the PI3K/AKT pathway in Parkinson's disease (PD) model mice. Methods The study explores the mitigating effects of Galangin on PD processes by administering 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to induce the condition. Techniques including network analysis, transcriptomic analysis, rotarod test, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, western blotting, immunohistochemistry, immunofluorescence, and hematoxylin-eosin (HE) were employed to unveil the molecular changes induced by Galangin. Results The network pharmacological analysis showed 301 targets related to Galangin, and 2,858 genes related to PD. Galangin treatment can improve the motor coordination of PD model mice, reduce damage to neurons in the brain, improve the antioxidant capacity and reduce the inflammatory damage of brain tissue. Additionally, Galangin suppressed mRNA expression of PD markers (IL-1β, TNF-α, IL-6, SRC and PTGS2), elevated protein levels of GSH-Px, SOD, P-PI3K, P-CREB, P-AKT, TH, BDNF and P62, while decreasing α-syn, SRC, MDA, Beclin-1 and LC3B expression. Moreover, the expression of significantly different genes in the Galangin-treated group and model group analyzed by transcriptomics was basically consistent with the qRT-PCR verification results. Conclusion Galangin supresses Beclin-1-dependent autophagy and upregulates the PI3K/AKT signaling pathway to attenuate the neuroinflammatory injury and improve motor coordination ability in PD mice induced by MPTP.
Collapse
Affiliation(s)
- Liping Huang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
- Mangrove Institute, Lingnan Normal University, Zhanjiang, China
| | - Qiaofeng Li
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Jingyi Wu
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Yingying He
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Junwei Huang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Sipeng Xie
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Canfeng Yang
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Qingling Ruan
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Zhongliu Zhou
- School of Chemistry and Chemical Engineering, Western Guangdong Characteristic Biomedical Engineering Technology Research Center, Lingnan Normal University, Zhanjiang, China
| | - Minzhen Deng
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ Department of Neurology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangzhou, China
| |
Collapse
|
13
|
Li X, Zeng Z. Exploring the link between estimated glucose disposal rate and Parkinson's disease: cross-sectional and mortality analysis of NHANES 2003-2016. Front Aging Neurosci 2025; 17:1548020. [PMID: 40256390 PMCID: PMC12006106 DOI: 10.3389/fnagi.2025.1548020] [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: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/22/2025] Open
Abstract
Objectives To investigate the association between estimated glucose disposal rate (eGDR), a surrogate marker of insulin resistance, and Parkinson's disease (PD) risk, and to examine the relationship between eGDR and all-cause mortality among PD patients. Methods Using data from the National Health and Nutrition Examination Survey (NHANES) 2003-2016, we conducted a cross-sectional study of 20,767 participants aged ≥40 years. eGDR was calculated using waist circumference, hypertension status, and HbA1c levels. PD cases were identified through anti-parkinsonian medication use. The association between eGDR and PD was examined using weighted logistic regression models with progressive adjustment for potential confounders. Survival analysis was performed in 255 PD patients to assess the relationship between eGDR and all-cause mortality. Results Among participants, 256 had PD (weighted prevalence: 1.23%). Higher eGDR was associated with lower odds of PD in crude analysis (OR: 0.906, 95% CI: 0.856-0.960, P < 0.001). After full adjustment, the highest eGDR tertile showed significantly lower odds of PD compared to the lowest tertile (OR: 0.574, 95% CI: 0.337-0.976, P = 0.040). Restricted cubic spline analysis revealed a significant M-shaped non-linear relationship between eGDR and PD risk (P for non-linearity < 0.001). In survival analysis, higher eGDR was associated with lower mortality risk (adjusted HR: 0.875, 95% CI: 0.775-0.987, P = 0.030), with an inverted U-shaped relationship observed (P for non-linearity = 0.0352). Conclusion Higher eGDR levels are associated with lower PD risk and better survival in PD patients, suggesting that insulin sensitivity might play a role in PD pathogenesis and progression. These findings highlight the potential importance of metabolic health in PD.
Collapse
Affiliation(s)
- Xiaoting Li
- Department of Neurology, Panyu Hexian Memorial Affiliated Hospital of Guangzhou, Guangzhou, China
| | - Zhaohao Zeng
- Department of Neurology, Shenzhen People’s Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
14
|
Shin SY, Kang IS, Kim C. ERK inhibits osteoclast differentiation in RAW 264.7 cells through the osteoprotegerin-mediated autophagy. Bone 2025; 193:117424. [PMID: 39947572 DOI: 10.1016/j.bone.2025.117424] [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: 11/05/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Osteoclasts (OCs) are bone-resorbing cells derived from the monocyte/macrophage lineage. The extracellular signal-regulated kinase (ERK) pathway controls cellular responses such as proliferation, differentiation, and survival, including those of OCs. In the present study, ERK inhibitors reduced the proliferation of bone marrow-derived macrophages (BMMs) and RAW 264.7 cells. However, ERK inhibitors decreased OC differentiation in BMMs but increased it in RAW 264.7 cells. ERK downregulation using small interfering RNA transfection also increased the OC differentiation and the expression of receptor activator of nuclear factor-κB, OC-specific markers, and OC-associated transcription factors in RAW 264.7 cells. These findings suggest ERK regulates OC differentiation in RAW 264.7 cells differently than in BMMs. Thus, we further investigated the mechanism by which ERK negatively regulates OC differentiation in RAW 264.7 cells. ERK inhibition decreased the expression of osteoprotegerin (OPG), a negative regulator of OC differentiation. OPG knockdown increased OC formation. ERK inhibitors activated the Akt/mammalian target of the rapamycin (mTOR) signaling pathway while inhibiting unc-51-like autophagy activating kinase 1 (ULK1). This resulted in decreased levels of microtubule-associated protein 1A/1B-light chain 3-II (LC3-II) and increased levels of p62, thereby reducing autophagy. In addition, OPG knockdown reduced autophagy by activating Akt/mTOR and inhibiting ULK1, resulting in decreased LC3-II and accumulated p62. Therefore, ERK inhibition promoted OC differentiation by downregulating OPG-mediated inhibition of osteoclastogenesis and autophagy in RAW 264.7 cells. These findings highlight ERK's complex role in OC differentiation and suggest that understanding ERK's dual impact on OC differentiation can provide insights into novel treatment strategies for bone-related disorders.
Collapse
Affiliation(s)
- Soo-Young Shin
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - In-Soon Kang
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Chaekyun Kim
- Laboratory of Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea; BK21, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
15
|
Xu Y, Liang H, Xia K, Yao J, Chen Y, Hou B, Hao Z. Umbelliferone attenuates calcium oxalate crystal-induced renal injury and inflammation by attenuating autophagy through the PI3K/AKT pathway. Int Immunopharmacol 2025; 150:114250. [PMID: 39952007 DOI: 10.1016/j.intimp.2025.114250] [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: 10/25/2024] [Revised: 01/14/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Calcium oxalate (CaOx) crystals are a major component of human kidney crystals and can induce renal tubular inflammation and damage, ultimately leading to renal calcium deposits and kidney stone formation. Umbelliferone (Umb) is a common coumarin compound. In this study, we used in vivo, in vitro experiments and network pharmacology were performed to assess the therapeutic effects of Umb on kidney stones and investigate its pharmacological mechanism. First, we established cellular and mouse models of calcium oxalate renal calcinosis, and we found that Umb reduces renal crystalline deposits, as well as the inflammation and damage they cause. Subsequently, we screened the PI3K/AKT signalling pathway via network pharmacology and experimentally demonstrated that Umb exerts its protective effects through the PI3K/AKT signalling pathway. Finally, molecular docking techniques and experiments were used to find out that Umb acts directly on PIK3CA to play its role.Our results indicate that Umb alleviates inflammation and injury by attenuating renal autophagy induced by kidney stones via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yuexian Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Hu Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Kaiguo Xia
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Junfeng Yao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China
| | - Yang Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| | - Bingbing Hou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| | - Zongyao Hao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, China.
| |
Collapse
|
16
|
Shi H, Yang Y, Gao J, Kumar S, Xie H, Chen Z, Lyu J, Sihto H, Koljonen V, Vega-Rubin-de-Celis S, Vukojevic V, Farnebo F, Björnhagen V, Höög A, Juhlin CC, Lee L, Wickström M, Becker JC, Johnsen JI, Larsson C, Lui WO. Kit-mediated autophagy suppression driven by a viral oncoprotein emerges as a crucial survival mechanism in Merkel cell carcinoma. Autophagy 2025:1-21. [PMID: 40108758 DOI: 10.1080/15548627.2025.2477385] [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: 03/17/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 03/22/2025] Open
Abstract
The KIT/c-KIT proto-oncogene is frequently over-expressed in Merkel cell carcinoma (MCC), an aggressive skin cancer commonly caused by Merkel cell polyomavirus (MCPyV). Here, we demonstrated that truncated MCPyV-encoded large T-antigen (LT) suppressed macroautophagy/autophagy by stabilizing and sequestering KIT in the paranuclear compartment via binding VPS39. KIT engaged with phosphorylated BECN1, thereby enhancing its association with BCL2 while diminishing its interaction with the PIK3C3 complex. This process ultimately resulted in the suppression of autophagy. Depletion of KIT triggered both autophagy and apoptosis, and decreased LT expression. Conversely, blocking autophagy in KIT-depleted cells restored LT levels and rescued apoptosis. Additionally, stimulating autophagy efficiently increased cell death and inhibited tumor growth of MCC xenografts in mice. These insights into the interplay between MCPyV LT and autophagy regulation reveal important mechanisms by which viral oncoproteins are essential for MCC cell viability. Thus, autophagy-inducing agents represent a therapeutic strategy in advanced MCPyV-associated MCC.Abbreviation: 3-MA, 3-methyladenine; AL, autolysosome; AP, autophagosome; Baf-A1, bafilomycin A1; BARA, β-α repeated autophagy specific domain; BH3, BCL2 homology 3 domain; CCD, coiled-coil domain; CHX, cycloheximide; Co-IP, co-immunoprecipitation; CQ, chloroquine; CTR, control; DAPI, 4',6-diamidino-2-phenylindole; EBSS, Earle's balanced salt solution; ECD, evolutionarily conserved domain; EEE, three-tyrosine phosphomimetic mutations Y229E Y233E Y352E; ER, endoplasmic reticulum; FFF, three-tyrosine non-phosphomimetic mutations; FFPE, formalin-fixed paraffin-embedded; FL, full-length; GIST, gastrointestinal stromal tumor; IB, immunoblotting; IHC, immunohistochemistry; KIT-HEK293, KIT stably expressing HEK293 cells; KRT20/CK20, keratin 20; LT, large T-antigen; LT339, MCPyV truncated LT antigen; LTco, codon-optimized MCPyV LT antigen; MCC, Merkel cell carcinoma; MCPyV-, MCPyV-negative; MCPyV, Merkel cell polyomavirus; MCPyV+, MCPyV-positive; PARP1, poly(ADP-ribose) polymerase 1; PCI, pan-caspase inhibitor; PI, propidium iodide; PtdIns3K, class III phosphatidylinositol 3-kinase; PtdIns3P, phosphatidylinositol-3-phosphate; RB1, RB transcriptional corepressor 1; RTKs, receptor tyrosine kinases; KITLG/SCF, KIT ligand; sT, small T-antigen; sTco, codon-optimized MCPyV sT antigen; T-B, Tat-BECN1; T-S, Tat-scrambled; TEM, transmission electron microscopy.
Collapse
Affiliation(s)
- Hao Shi
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Yajie Yang
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Jiwei Gao
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Satendra Kumar
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Hong Xie
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Jiawen Lyu
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Harri Sihto
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Vladana Vukojevic
- Laboratory of Experimental Alcohol and Drug Addiction Research, Department of Clinical Neuroscience, Karolinska Institutet; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Filip Farnebo
- Department of Molecular Medicine and Surgery, Karolinska Institutet; Clinic for Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Viveca Björnhagen
- Department of Molecular Medicine and Surgery, Karolinska Institutet; Clinic for Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Höög
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Linkiat Lee
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Malin Wickström
- Department of Women's and Children's Health, Karolinska Institutet; BioClinicum J5:20, Karolinska University Hospital, Solna, Sweden
| | - Jürgen C Becker
- Translational Skin Cancer Research, University Medicine Essen, Essen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Institute (DKFZ), Heidelberg, Baden-Württemberg, Germany
| | - John Inge Johnsen
- Department of Women's and Children's Health, Karolinska Institutet; BioClinicum J5:20, Karolinska University Hospital, Solna, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| | - Weng-Onn Lui
- Department of Oncology-Pathology, Karolinska Institutet; BioClinicum J6:20, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
17
|
Dhapola R, Kumari S, Sharma P, Vellingiri B, HariKrishnaReddy D. Advancements in autophagy perturbations in Alzheimer's disease: Molecular aspects and therapeutics. Brain Res 2025; 1851:149494. [PMID: 39922409 DOI: 10.1016/j.brainres.2025.149494] [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: 09/23/2024] [Revised: 12/11/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Emerging evidences suggest that autophagy, a key cellular process responsible for degrading and recycling damaged organelles and proteins, plays a crucial role in maintaining neuronal health. Dysfunctional autophagy has been linked to the pathogenesis of Alzheimer's disease (AD), contributing to the accumulation of misfolded proteins and cellular debris. Molecular mechanisms underlying autophagy dysfunction in AD involve amyloid-beta (Aβ) and tau accumulation, neuroinflammation, mitochondrial dysfunction, oxidative stress and endoplasmic reticulum stress. Disrupted signaling pathways such as TRIB3, Nmnat and BAG3 that regulate key processes like autophagosome initiation, lysosome function, and protein homeostasis also play a crucial role in the pathogenesis. Restoration of autophagy by modulating these molecular and signaling pathways may be an effective therapeutic strategy for AD. Studies have found few drugs targeting autophagy dysregulation in AD. These drugs include metformin that has been found to modulate the expression of TRIB3 for autophagy regulation. Another drug, resveratrol has been reported to augment the activity of Nmnat thus, increases autophagy flux. BACE1 and mTOR inhibitors like arctigenin, nilvadipine and dapagliflozin were also found to restore autophagy. This study elaborates recent advances in signaling and molecular pathways and discusses current and emerging therapeutic interventions targeting autophagy dysfunction in AD.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab 151401 Bathinda, Punjab, India.
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401 Punjab, India.
| |
Collapse
|
18
|
Wang J, Ren W, Sun Z, Liu S, Han Z, Wang Y, Zeng Y, Meng J, Yao X. Impact of intragastric administration of donkey milk on mouse immunity utilizing gut microbiomics and plasma metabolomics. Front Vet Sci 2025; 12:1486406. [PMID: 40144527 PMCID: PMC11938429 DOI: 10.3389/fvets.2025.1486406] [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: 08/26/2024] [Accepted: 02/17/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Donkey milk demonstrates closer compositional resemblance to human milk compared to bovine milk, positioning it as an optimal nutritional substitute for infants with cow's milk allergy. Furthermore, its rich profile of bioactive compounds suggests potential immunomodulatory properties. This study systematically investigated the effects of donkey milk supplementation on murine immune function and gut microbiome dynamics, thereby providing mechanistic insights to support its clinical development in functional food applications. Methods Following daily intragastric administration of 10 mL/kg of body weight of donkey milk (DM) or distilled water (DW) to the mice for 28 consecutive days, liver tissues were harvested for immunological profiling, with concurrent collection of blood samples for plasma metabolomic analysis and fecal specimens for gut microbiome characterization. Subsequently, the modulatory effects of donkey milk supplementation on immune parameters, intestinal microbiota composition, and plasma metabolic profiles were systematically evaluated. Results Immunity analysis revealed that intragastric administration of DM raised the levels of IL-6 and TNF-α cytokines in mouse liver. In addition, DM modulated the composition of both the murine gut microbiome and plasma metabolites. One-hundred and forty-five differentially-produced metabolites were identified, most prominently nicotinamide, L-valine, and β-estradiol, that are primarily associated with valine, leucine, and isoleucine biosynthesis and degradation, nicotinate and nicotinamide metabolism, and unsaturated fatty acid biosynthesis. Alterations at phylum, genus, and species levels were evident in the fecal microbiota of mice after intragastric administration of DM. In particular, an increased abundance of the Lactobacillus bacterium was observed. Correlation analysis of differential metabolites and microbiomes indicated a correspondence between Falsiroseomonas and Salipiger species and the antioxidant coenzyme Q that has the potential to activate the immune system. Conclusion The data collectively suggest that DM may adjust the murine gut microbiome and plasma metabolites thereby potentially improving immunity in mice.
Collapse
Affiliation(s)
- Jianwen Wang
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
- Xinjiang Key Laboratory of Equine Breeding and Exercise Physiology, Ürümqi, China
| | - Wanlu Ren
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
| | - Zhiwen Sun
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
| | - Shibo Liu
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
| | - Zixiang Han
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
| | - Yongfa Wang
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
| | - Yaqi Zeng
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
- Xinjiang Key Laboratory of Equine Breeding and Exercise Physiology, Ürümqi, China
| | - Jun Meng
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
- Xinjiang Key Laboratory of Equine Breeding and Exercise Physiology, Ürümqi, China
| | - Xinkui Yao
- College of Animal Science, Xinjiang Agricultural University, Ürümqi, China
- Xinjiang Key Laboratory of Equine Breeding and Exercise Physiology, Ürümqi, China
| |
Collapse
|
19
|
Long S, Wang Y. Association of TAB2 gene polymorphism with endometrial cancer susceptibility and clinical analysis. Turk J Obstet Gynecol 2025; 22:1-12. [PMID: 40062608 PMCID: PMC11894771 DOI: 10.4274/tjod.galenos.2025.24983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 03/14/2025] Open
Abstract
Objective Transforming growth factor-β-activated kinase 1 binding protein 2 (TAB2) plays a vital role in inflammatory pathways. It has also been considered a potential target for the enhancement of the the antiestrogen effects. Previous evidence has indicated that TAB2 gene variants are associated with several diseases, whereas their potential correlation with endometrial cancer (EC) is unclear. This study aims to initially explore the association between TAB2 gene polymorphisms (rs237028 /AG, rs521845 T/G, and rs652921 T/C) and EC. Materials and Methods Polymerase chain reaction-restriction fragment length polymorphism was applied to determine the genotype composition and the allele frequencies of TAB2 gene variant polymorphisms in 270 EC patients and 294 healthy controls. Results The G allele of rs521845 was related to the increase of EC risk [p=0.08, odds ratio (OR): 0.72, 95% confidence interval (CI): 0.56-0.91]. Moreover, EC risk was associated with rs521845 in different genetic models (p=0.017, OR: 0.63, 95% CI: 0.44-0.91 in the codominant model; p=0.0051, OR: 0.61, 95% CI: 0.43-0.87 in the dominant model). For rs237028, the percentage of AG genotype in patients with highly differentiated tumours (G1) was significantly higher than that in moderately, poorly differentiated patients (G2/G3) (p=0.031, OR: 0.77, 95% CI: 0.45-1.30). Conclusion Our results showed that the rs521845 polymorphism of TAB2, was associated with EC risk, suggesting that TAB2 may play a crucial role in EC prognosis.
Collapse
Affiliation(s)
- Siyu Long
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
- Sichuan University West China Second University Hospital, Clinic of Andrology/Sichuan Human Sperm Bank, Chengdu, China
| | - Yanyun Wang
- Sichuan University, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Center for Translational Medicine, Laboratory of Molecular Translational Medicine, Sichuan, China
| |
Collapse
|
20
|
Jiang Y, Zhang H, Shi J, Shan T, Liu M, Wang P, Liang X, Liang H. Nicotinamide riboside alleviates sweeteners-induced brain and cognitive impairments in immature mice. Food Funct 2025; 16:1947-1968. [PMID: 39957299 DOI: 10.1039/d4fo05553e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The consumption of sweeteners is high around the world. Sweet beverages are one of the most important and popular sources of sweeteners. Previous studies have reported that excessive sweeteners might cause health hazards, including cognitive impairment. Nicotinamide riboside (NR), a precursor of NAD+, has been found to alleviate several cognitive impairments. However, the protective effects of NR against sweetener-induced cognitive impairment remain unclear. Hence, we evaluated the effects of sweeteners and NR (400 mg kg-1 d-1) on the brain and cognition of mice by simulating an extreme lifestyle of completely replacing water with sugar-sweetened beverage (simulated with 10% sucrose solution) or sugar-free sweet beverage (simulated with 0.05% aspartame solution) from weaning to adulthood. The results revealed that continuous exposure to sucrose or aspartame for eight weeks did not significantly cause differences in body weight but significantly induced cognitive impairments, including anxiety- and depressive-like behaviours, impairments in learning, memory and sociability. Moreover, sucrose or aspartame exposure induced neuronal injury, reduction of Nissl bodies, overactivation of the TLR4/NF-κB/NLRP3/ASC/Caspase-1 pathway and increased downstream inflammatory cytokines in mouse hippocampus, and also induced an imbalance of oxidative stress, apoptosis and autophagy, large consumptions of intracellular antioxidant factors, and overactivation of the PI3K/Akt/FOXO1 and PI3K/Akt/mTOR pathways in mouse brain. NR treatment increased NAD+ in the brain, and prevented and alleviated these impairments effectively. In summary, we found that NR supplementation protected against cognitive impairment caused by sucrose or aspartame in immature mice, which might be related to increased brain NAD+ level, relieved neuroinflammation and pyroptosis in the hippocampus, and maintained a balance of oxidative stress, apoptosis and autophagy in the brain.
Collapse
Affiliation(s)
- Yushan Jiang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Jing Shi
- College of continuing education, Qingdao University, Qingdao, China
| | - Tianhu Shan
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Man Liu
- Basic Medical College, Qingdao University, Qingdao, China
| | - Peng Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Xi Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Ma Y, Ma Y, Li P, Ma F, Yu M, Xu J, Yang Y. Wnt5a alleviates the symptoms of PCOS by modulating PI3K/AKT/mTOR pathway-mediated autophagy in granulosa cells. Cell Signal 2025; 127:111575. [PMID: 39710088 DOI: 10.1016/j.cellsig.2024.111575] [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: 10/17/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) is a metabolic and endocrine disease that entails dysregulated ovulation, hyperandrogenism, and polycystic ovaries. While Wnt5a has been suggested to play key roles in follicular development and female fertility under normal conditions, its functions in the context of PCOS have yet to be established. This study was thus designed to explore the impact of Wnt5a on ovarian granulosa cell autophagy in PCOS, providing in vitro evidence in support of its role in this setting. METHODS DHT-induced granulosa (KGN) cells were used as an in vitro model, and Wnt5a and autophagy-related protein levels in these cells were detected via Western blotting. Downregulating the expression of Wnt5a in KGN cells (by interference and inhibitor) was also performed, and Western blotting, RT-PCR, and immunofluorescence strategies were used to detect autophagy-related and PI3K/AKT/mTOR pathway-associated factors in this setting. In vivo, BOX5 was tested as a therapeutic inhibitor of Wnt5a in a murine model of DHEA-induced PCOS. Changes in ovarian morphology were detected through hematoxylin staining, while E2 and T hormone levels were quantified by ELISA, and autophagy-related factors in these animals were quantified through Western blotting, immunofluorescence, and immunohistochemistry. RESULTS Wnt5a and autophagy-related protein levels rose significantly in DHT-induced KGN cells. Following downregulation of the Wnt5a in these cells, a significant decrease in autophagy-related factor levels was noted relative to the DHT group, together with significant increases in pathway-related factors. In mice, BOX5 treatment was sufficient to restore serum levels of androgen and to improve polycystic ovarian changes, while also suppressing the levels of autophagy-associated factors within ovarian granulosa cells. CONCLUSION Wnt5a downregulation suppresses autophagy in PCOS granulosa cells through the activation of the PI3K/AKT/mTOR pathway, in addition to remediating polycystic ovarian changes and normalizing serum levels of sex hormones.
Collapse
Affiliation(s)
- Yabo Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Yuqin Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Pengfei Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Fucheng Ma
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Miao Yu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Jinrui Xu
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China
| | - Yi Yang
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan 750021, China; School of Life Sciences, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
22
|
Di-Luoffo M, Schmitter C, Barrere EC, Therville N, Chaouki M, D'Angelo R, Arcucci S, Thibault B, Delarue M, Guillermet-Guibert J. Mechanical compressive forces increase PI3K output signaling in breast and pancreatic cancer cells. Life Sci Alliance 2025; 8:e202402854. [PMID: 39746759 PMCID: PMC11707390 DOI: 10.26508/lsa.202402854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Mechanical stresses, including compression, arise during cancer progression. In solid cancer, especially breast and pancreatic cancers, the rapid tumor growth and the environment remodeling explain their high intensity of compressive forces. However, the sensitivity of compressed cells to targeted therapies remains poorly known. In breast and pancreatic cancer cells, pharmacological PI3K inactivation decreased cell number and induced apoptosis. These effects were accentuated when we applied 2D compression forces in mechanically responsive cells. Compression selectively induced the overexpression of PI3K isoforms and PI3K/AKT pathway activation. Furthermore, transcriptional effects of PI3K inhibition and compression converged to control the expression of an autophagy regulator, GABARAP, whose level was inversely associated with PI3K inhibitor sensitivity under compression. Compression alone blocked autophagy flux in all tested cells, whereas inactivation of basal PI3K activity restored autophagy flux only in mechanically non-responsive compressed cells. This study provides direct evidence for the role of the PI3K/AKT pathway in compression-induced mechanotransduction. PI3K inhibition promotes apoptosis or autophagy, explaining PI3K importance to control cancer cell survival under compression.
Collapse
Affiliation(s)
- Mickaël Di-Luoffo
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Céline Schmitter
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Emma C Barrere
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Nicole Therville
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Maria Chaouki
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Romina D'Angelo
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Silvia Arcucci
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Benoit Thibault
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
| | - Morgan Delarue
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Julie Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Labex Toucan, Toulouse, France
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| |
Collapse
|
23
|
Abdelsalam RM, Hamam HW, Eissa NM, El-Sahar AE, Essam RM. Empagliflozin Dampens Doxorubicin-Induced Chemobrain in Rats: The Possible Involvement of Oxidative Stress and PI3K/Akt/mTOR/NF-κB/TNF-α Signaling Pathways. Mol Neurobiol 2025; 62:3480-3492. [PMID: 39302617 DOI: 10.1007/s12035-024-04499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Chemobrain is a cognitive impairment observed in up to 75% of cancer patients treated with doxorubicin (DOX). Cognitive deficits associated with DOX are complex, and multiple interplay pathways contribute to memory impairment and the loss of concentration. Empagliflozin (EMPA), a sodium-glucose co-transporter-2 (SGLT-2) inhibitor with neuroprotective potential, has recently been elucidated because of its regulatory effects on oxidative stress and neuroinflammation. Thus, this study aimed to explore the protective mechanisms of EMPA in DOX-induced chemobrain. Rats were allocated to four groups: normal (NC), EMPA, DOX, and EMPA + DOX. Chemobrain was induced in the third and fourth groups by DOX (2 mg/kg, IP) on the 0th, 7th, 14th, and 21st days of the study, while EMPA was administered (10 mg/kg, PO) for 28 consecutive days in both the EMPA and EMPA + DOX groups. Behavioral and biochemical assessments were then performed. Rats treated with DOX exhibited significant memory, learning, and muscle coordination dysfunctions. Moreover, DOX boosted oxidative stress in the brain, as evidenced by elevated malondialdehyde (MDA) content together with decreased levels of nuclear factor-erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) and reduced glutathione (GSH). Neuroinflammation was also observed as an upsurge of tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB) (p65). Additionally, DOX diminished the expression of brain-derived neurotrophic factor (BDNF) and increased phosphoinositol-3-kinase (PI3K), phosphorylated-Akt (pAkt), and mammalian target of rapamycin (mTOR) content. EMPA exhibited potent neuroprotective potential in DOX-induced cognitive impairment, attributed to its antioxidant and neuroplasticity-enhancing properties and suppression of the PI3K/Akt/mTOR/NF-κB/TNF-α signaling pathway.
Collapse
Affiliation(s)
- Rania M Abdelsalam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt
| | - Hatem W Hamam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Noha M Eissa
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Ayman E El-Sahar
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt
| | - Reham M Essam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt.
| |
Collapse
|
24
|
Wang Z, Chen H, Xiong S, Chen X, Gao X, Huang P, Zou J, Cao H. Lactobacillus plantarum SMUM211204 Exopolysaccharides Have Tumor-Suppressive Effects on Colorectal Cancer by Regulating Autophagy via the mTOR Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40017402 DOI: 10.1021/acs.jafc.4c09818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Probiotics have demonstrated their ability to suppress tumors in cell lines and mouse models. However, the precise molecules responsible for these effects remain unidentified. We focused on isolating and analyzing the exopolysaccharides (EPSs) produced by Lactobacillus plantarum (L. plantarum) SMUM211204. Our findings confirm that EPSs impair the growth of HCT116 cells and induce autophagy and apoptosis. Moreover, further experimental evidence demonstrates that EPSs diminish the expression of phosphorylation levels of PI3K, AKT, and mTOR. In contrast, they boost the expression of AMPKa, elevate the ULK1 level, and increase the protein LC3-II/I ratio. Furthermore, when rapamycin is employed to impede EPS-induced autophagy, it results in an enhancement of apoptosis and cell death in HCT116 cells. To validate these findings in vivo, we conducted an animal study using a colorectal cancer xenograft model. The results showed a significant reduction in tumor volume and weight in the EPS-treated group compared with the control group. Immunohistochemical analysis of tumors indicated increased expressions of LC3 and caspase-3, along with decreased levels of phospho-PI3Kinase, phospho-AKT, and P62, consistent with in vitro findings. Our study proved that EPSs have an inhibitory effect on colorectal cancer and can be used as a preventive and therapeutic drug for cancer.
Collapse
Affiliation(s)
- Zixuan Wang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hengqiu Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Shasha Xiong
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaoliang Chen
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xuefeng Gao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Pengwei Huang
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jinhu Zou
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Hong Cao
- Department of Microbiology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
25
|
Bao Y, Sun C. Investigating the Role of Buzhong Yiqi Decoction on Neurogenic Bladder with Network Pharmacology, Molecular Docking, and In Vitro Assays. Assay Drug Dev Technol 2025. [PMID: 39957354 DOI: 10.1089/adt.2024.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2025] Open
Abstract
Buzhong Yiqi decoction (BZYQD) is a traditional Chinese medicine prescription for treating neurogenic bladder (NB). However, the underlying pharmacological mechanism remains unclear. This study aims to clarify the related molecular mechanism. Molecular structure information and targets of core components of BZYQD were obtained from Traditional Chinese Medicines Systems Pharmacology Platform (TCMSP) and SwissTargetPrediction databases. Genes involved in NB were obtained from Comparative Toxicogenomics Database, DisGeNet, GeneCards, and Online Mendelian Inheritance in Man databases. The hub targets of BZYQD in NB treatment were identified by protein-protein interaction (PPI) network analysis with STRING platform and analyzed by gene ontology analysis and the Kyoto Encyclopedia of Genes and Genomics pathway enrichment analysis. Molecular docking was used to verify the binding affinity between the hub targets and the bioactive components of BZYQD. Subsequently, the neuroprotective and anti-inflammatory effects of main bioactive components of BZYQD were investigated with in vitro assays. A total of 131 candidate compounds and 925 predicted target genes were screened. PPI network analysis suggested that ESR1, EGFR, HSP90AA1, MAPK3, AKT1, and CASP3 were the hub targets. BZYQD treatment was associated with hypoxia inducible factor-1 (HIF-1) signaling pathway. Dehydroglyasperin C (DGC), N-cis-feruloyltyramine, shinpterocarpin (SHI), gancaonin M, and glyasperin B, as the main bioactive components of BZYQD, had good binding affinity with hub target proteins. DGC and SHI treatment could significantly inhibit the injury of neurons and inflammatory response of microglia stimulated by oxidized low-density lipoprotein (ox-LDL), respectively. In summary, BZYQD and its main bioactive components DGC and SHI show good potential to ameliorate the symptoms of NB.
Collapse
Affiliation(s)
- Yixin Bao
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chun Sun
- Department of Urology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
26
|
Tao D, Li F, Zhang X, Guo H, Yang R, Yang Y, Zhang L, Shen Z, Teng J, Chen P, He B. 20(R)-ginsenoside Rg3 protects against focal cerebral ischemia‒reperfusion injury by suppressing autophagy via PI3K/Akt/mTOR signaling pathway. Neuropharmacology 2025; 263:110226. [PMID: 39557153 DOI: 10.1016/j.neuropharm.2024.110226] [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: 07/08/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE This study aimed to investigate the effect of 20(R)-ginsenoside Rg3 on autophagy induced by cerebral ischemia‒reperfusion injury (CIRI) in rats and explore its regulation of the PI3K/Akt signaling pathway. METHODS Middle cerebral artery occlusion/reperfusion (MCAO/R) in male rats was injected intraperitoneally with 20(R)-ginsenoside Rg3 (5, 10, 20 mg/kg) 12 h before modeling, 2 h after ischemia and 12 h after reperfusion. Neurobehavioral and neuronal morphological changes were detected 24 h after brain I/R. In vitro, the OGD/R-induced injury model is replicated in PC12 cells and different concentrations of 20(R)-ginsenoside Rg3 are administered to observe its effects on cell viability and autophagy and PI3K/Akt/mTOR-related protein expression. RESULTS Our findings suggest that treatment with 20 mg/kg 20(R)-ginsenoside Rg3 significantly attenuated the neuronal injury, as evidenced by a decreased number of damaged neurons, reduced dissolution of Nissl corpuscles, a fewer autophagosomes, and downregulated expression of Beclin1 and LC3-II/I compared with the MCAO/R group. Furthermore, 20(R)-ginsenoside Rg3 treatment significantly upregulated the expression of p62, p-PI3K, p-AKT, and p-mTOR. In vitro, 20(R)-ginsenoside Rg3 significantly improved the survival rate of cells following OGD/R and markedly attenuated the LY294002 and OGD/R-induced upregulation of Beclin1 and LC3 gene expression. Moreover, 20(R)-ginsenoside Rg3 could rescued the LY294002 and OGD/R-induced downregulation of p62, p-PI3K, p-AKT, and p-mTOR expression. CONCLUSIONS 20(R)-ginsenoside Rg3 attenuates neuronal injury and motor dysfunction following ischemia-reperfusion by inhibiting the activation of autophagy, and its mechanism is related to the upregulation of the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Daiju Tao
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Fajing Li
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China; The First People's Hospital of Liangshan Yi Autonomous Prefecture, XiChang, Sichuan Province, 615000, PR China
| | - Xiaochao Zhang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Hui Guo
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China; Department of Pharmacology, Haiyuan College, Kunming Medical University, 650106, PR China
| | - Renhua Yang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Yuan Yang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Li Zhang
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Zhiqiang Shen
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China
| | - Jia Teng
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China; Department of Pharmacology, Haiyuan College, Kunming Medical University, 650106, PR China.
| | - Peng Chen
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China.
| | - Bo He
- School of Pharmaceutical Science, Yunnan Key Laboratory of Pharmacology for Natural Products, NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, 650500, PR China.
| |
Collapse
|
27
|
Bao Y, Ma Y, Huang W, Bai Y, Gao S, Xiu L, Xie Y, Wan X, Shan S, Chen C, Qu L. Regulation of autophagy and cellular signaling through non-histone protein methylation. Int J Biol Macromol 2025; 291:139057. [PMID: 39710032 DOI: 10.1016/j.ijbiomac.2024.139057] [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: 09/17/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Autophagy is a highly conserved catabolic pathway that is precisely regulated and plays a significant role in maintaining cellular metabolic balance and intracellular homeostasis. Abnormal autophagy is directly linked to the development of various diseases, particularly immune disorders, neurodegenerative conditions, and tumors. The precise regulation of proteins is crucial for proper cellular function, and post-translational modifications (PTMs) are key epigenetic mechanisms in the regulation of numerous biological processes. Multiple proteins undergo PTMs that influence autophagy regulation. Methylation modifications on non-histone lysine and arginine residues have been identified as common PTMs critical to various life processes. This paper focused on the regulatory effects of non-histone methylation modifications on autophagy, summarizing related research on signaling pathways involved in autophagy-related non-histone methylation, and discussing current challenges and clinical significance. Our review concludes that non-histone methylation plays a pivotal role in the regulation of autophagy and its associated signaling pathways. Targeting non-histone methylation offers a promising strategy for therapeutic interventions in diseases related to autophagy dysfunction, such as cancer and neurodegenerative disorders. These findings provide a theoretical basis for the development of non-histone-methylation-targeted drugs for clinical use.
Collapse
Affiliation(s)
- Yongfen Bao
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Yaoyao Ma
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China
| | - Wentao Huang
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410013, China
| | - Yujie Bai
- Department of Scientific Research and Education, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang 330000, China
| | - Siying Gao
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Luyao Xiu
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuyang Xie
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinrong Wan
- Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shigang Shan
- School of Public Health and Nursing, Hubei University of Science and Technology, Hubei 437000, China
| | - Chao Chen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lihua Qu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning 437000, China; School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, China.
| |
Collapse
|
28
|
Qin XD, Liang JF, Gan LY, Peng KS, Huang XH, Li XT, Chen JL, Li W, Zhang L, Jian J, Lu J. Blockage of polycystin-2 alleviates myocardial ischemia/reperfusion injury by inhibiting autophagy through the Ca 2+/Akt/Beclin 1 pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119892. [PMID: 39689827 DOI: 10.1016/j.bbamcr.2024.119892] [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: 07/07/2024] [Revised: 11/19/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
Autophagy is a well-conserved self-protection process that plays an important role in cardiovascular diseases. Excessive autophagy during myocardial ischemia/reperfusion injury (MIRI) induces calcium overload and the overactivation of an autophagic response, thereby aggravating cardiomyocyte damage. Polycystin-2 (PC2) is a Ca2+-permeable nonselective cation channel implicated in the regulation of autophagy. In the present study, autophagy was upregulated in myocardial ischemia/reperfusion in vivo and in vitro. PC2 knockdown using adeno-associated virus 9 particles containing Pkd2 short hairpin RNA infection markedly ameliorated MIRI, evidenced by reduced infarct size, diminished morphological changes, decreased cTnI levels, and improved cardiac function. Silencing PC2 reduced the autophagic flux in H9c2 cells. PC2 overexpression-mediated autophagic flux was inhibited by intracellular Ca2+ chelation with BAPTA-AM. Furthermore, PC2 ablation upregulated p-Akt (Ser473) and downregulated Beclin 1 in H/R. BAPTA-AM downregulated p-Akt(Ser473) and upregulated Beclin 1in PC2-overexpressing H9c2 cells. Moreover, the Akt inhibitor MK2206 abolished the BAPTA-AM-blunted PC2-dependent control of autophagy. Collectively, these results indicated that blockade of PC2 may be associated with the Ca2+/Akt/Beclin 1 signaling, thereby inhibiting excessive autophagy and serving as a potential strategy for mitigating MIRI.
Collapse
Affiliation(s)
- Xiao-Dan Qin
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jian-Feng Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Chinese People's Liberation Army Joint Logistic Support Force Lushan Rehabilitation and Recuperation Center, Jiujiang 332000, China
| | - Lin-Yu Gan
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Faculty of Pharmacy, Guiping People's Hospital, Guiping 537200, China
| | - Ke-Shan Peng
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Xue-Hong Huang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Xiao-Ting Li
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Jin-Li Chen
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Wan Li
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Lei Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Guilin Medical University, Guilin 541004, China
| | - Jie Jian
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China
| | - Jun Lu
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
29
|
Jiang M, Chi J, Qiao Y, Wang J, Zhang Z, Liu J, Sheng X, Yuan L. Ginsenosides Rg1, Rb1 and rare ginsenosides: Promising candidate agents for Parkinson's disease and Alzheimer's disease and network pharmacology analysis. Pharmacol Res 2025; 212:107578. [PMID: 39756554 DOI: 10.1016/j.phrs.2025.107578] [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: 10/12/2024] [Revised: 12/12/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
Ginseng has been commonly used as a traditional Chinese medicine in Asian countries for thousands of years. Ginsenosides are the main pharmacologically active ingredients isolated from ginseng and have neuroprotective effects in the treatment of neurodegenerative disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To summarise and investigate the protective roles of ginsenosides and their underlying mechanisms in PD and AD, we used ''Ginsenoside", ''Parkinson's disease", ''Alzheimer's disease", ''anti-inflammatory", ''antioxidant", and ''apoptosis" as keywords to search and extract relevant literature information from scientific databases such as Elsevier, PubMed, and Google Scholar databases. In particular, we used network pharmacology to identify the potential targets of ginsenosides Rg1 and Rb1 in PD and AD. By analysing the existing research advances and network pharmacology results, we found that the neuroprotective effects of ginsenosides, primarily mediated through anti-inflammation, anti-apoptosis and anti-oxidative stress, etc, may be associated with the PI3K/Akt, BDNF/TrkB, MAPKs, NF-κB, Nrf2 and Wnt/β-catenin signalling pathways. This review systematically summarises the different roles and mechanisms of ginsenosides Rg1, Rb1, and rare ginsenosides in PD and AD and provides new strategies for the treatment of neurodegenerative disorders. Network pharmacology provides a new research paradigm for the treatment of PD and AD using Rg1 and Rb1.
Collapse
Affiliation(s)
- Mingchun Jiang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China
| | - Jiaxin Chi
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Yifan Qiao
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jinpeng Wang
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Zhixin Zhang
- School of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jia Liu
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xinhao Sheng
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Liangjie Yuan
- School of Clinical Medicine and Basic Medical Science, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China; The Second Affiliated Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271000, China.
| |
Collapse
|
30
|
Ahmadzadeh AM, Pourbagher-Shahri AM, Forouzanfar F. Neuroprotective effects of phytochemicals through autophagy modulation in ischemic stroke. Inflammopharmacology 2025; 33:729-757. [PMID: 39884996 DOI: 10.1007/s10787-024-01606-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/14/2024] [Indexed: 02/01/2025]
Abstract
Stroke is a serious life-threatening medical condition. Understanding the underlying molecular mechanisms of this condition is crucial to identifying novel therapeutic targets that can improve patient outcomes. Autophagy is an essential mechanism for the destruction of damaged intracellular components that maintains homeostasis in physiological or pathological conditions. This process is involved in the pathophysiology of stroke. Phytochemicals are bioactive naturally occurring compounds present in plants. This paper reviews the neuroprotective roles of phytochemicals in ischemic stroke through autophagy modulation. It summarizes the interactions of various phytochemicals with key molecular targets of the autophagy pathway in ischemic stroke, including PI3K/Akt/mTOR, Beclin-1, and AMPK. Due to the ability of various phytochemicals to alter autophagic flux, they may provide promising opportunities in the development of new treatments and the improvement of stroke management.
Collapse
Affiliation(s)
- Amir Mahmoud Ahmadzadeh
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Radiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fatemeh Forouzanfar
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
31
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [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: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
32
|
Baazaoui N, Y Alfaifi M, Ben Saad R, Garzoli S. Potential role of long noncoding RNA maternally expressed gene 3 (MEG3) in the process of neurodegeneration. Neuroscience 2025; 565:487-498. [PMID: 39675694 DOI: 10.1016/j.neuroscience.2024.12.023] [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: 03/24/2024] [Revised: 10/28/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Neurodegenerative diseases (ND) are complex diseases of still unknown etiology. Lately, long non-coding RNAs (lncRNAs) have become increasingly popular and implicated in several pathologies as they have several roles and appear to be involved in all biological processes such as cell signaling and cycle control as well as translation and transcription. MEG3 is one of these and acts by binding proteins or directly or competitively binding miRNAs. It has a crucial role in controlling cell death, inflammatory process, oxidative stress, endoplasmic reticulum stress, epithelial-mesenchymal transition and other processes. Recent reports showed that MEG3 is a major driving force of the necrosis phenomena in AD, causing the death of neurons, and its upregulation in cancer patients was linked to tumor suppression. Dysregulation of MEG3 affects neuronal cell death, inflammatory process, smooth muscle cell proliferation and consequently leads to the initiation or the acceleration of the disease. This review examines the current state of knowledge concerning the level of expression and the regulatory function of MEG3 in relation to several NDs. In addition, we examined the relation of MEG3 with neurotrophic factors such as Tumor growth factor β (TGFβ) and its possible mechanism of action. A comprehensive and in-depth analysis of the role of MEG3 in ND could give a clearer picture about the initiation of the process of neuronal death and help develop an alternative therapy that targets MEG3.
Collapse
Affiliation(s)
- Narjes Baazaoui
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia
| | - Mohammad Y Alfaifi
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia; Tissue Culture and Cancer Biology Research Laboratory, King Khalid University, Abha 9004, Saudi Arabia
| | - Rania Ben Saad
- Biotechnology and Plant Improvement Laboratory, Center of Biotechnology of Sfax, B.P "1177", Sfax 3018, Tunisia
| | - Stefania Garzoli
- Department of Chemistry and Technologies of Drug, Sapienza University, P. le Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
33
|
Lv Q, Xu W, Yang F, Wei W, Chen X, Zhang Z, Liu Y. Reproductive Toxicity of Zearalenone and Its Molecular Mechanisms: A Review. Molecules 2025; 30:505. [PMID: 39942610 PMCID: PMC11821083 DOI: 10.3390/molecules30030505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Zearalenone (ZEA) is one of the common mycotoxins in feeds. ZEA and its metabolites have estrogen-like activity and can competitively bind to estrogen receptors, causing reproductive dysfunction and damage to reproductive organs. The toxicity mechanism of ZEA mainly inhibits the antioxidant pathway and antioxidant enzyme activity, induces cell cycle arrest and DNA damage, and blocks the process of cellular autophagy to produce toxic effects. In animal husbandry practice, when animals ingest ZEA-contaminated feed, it is likely to lead to abortion in females, abnormal sperm viability in males with inflammatory reactions in various organs, and cancerous changes in the reproductive organs of humans when they ingest contaminated animal products. In this paper, we reviewed in detail how ZEA induces oxidative damage by inducing the generation of reactive oxygen species (ROS) and regulating the expression of genes related to oxidative pathways, induces germ cell apoptosis through the mitochondrial and death receptor pathways, and activates the expression of genes related to autophagy in order to induce cellular autophagy. In addition, the molecular detoxification mechanism of ZEA is also explored in this paper, aiming to provide a new direction and theoretical basis for the development of new ZEA detoxification methods to better reduce the global pollution and harm caused by ZEA.
Collapse
Affiliation(s)
- Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, No. 263, Kaiyuan Avenue, Luoyang 471023, China; (W.X.); (W.W.)
| | | | | | | | | | | | | |
Collapse
|
34
|
Jin S, Liu X, Cai L, Yan J, Li L, Dong H, Gao Y, Zhu X, Zhang C, Xu X. Itraconazole promotes melanoma cells apoptosis via inhibiting hedgehog signaling pathway-mediated autophagy. Front Pharmacol 2025; 16:1545243. [PMID: 39917616 PMCID: PMC11798931 DOI: 10.3389/fphar.2025.1545243] [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: 12/14/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Background Itraconazole, a widely used antifungal medication, has shown potential in inhibiting tumor growth and reducing angiogenesis. However, its role in melanoma tumor growth remains insufficiently explored. This study investigates the inductive effect of itraconazole on autophagy-mediated apoptosis in melanoma cells. Method Potential drug targets were identified using the PMF machine learning algorithm. Apoptosis and cell cycle in melanoma cell lines A375 and A2058 were assessed via flow cytometry. Western blot analysis was performed to examine autophagy and associated signaling proteins, while autophagy flux and autophagosome formation were visualized using fluorescence microscopy. A melanoma cell xenograft mouse model was established to evaluate the inhibitory mechanisms of itraconazole on tumor cell proliferation. Result Using the PMF machine learning algorithm, SQSTM1 was identified as the primary target of itraconazole. Itraconazole inhibited melanoma cell proliferation by inducing G1 phase arrest and autophagy-mediated apoptosis in A375 and A2058 cells. Furthermore, itraconazole suppressed Hedgehog signaling and counteracted the activation of the Hedgehog agonist recombinant human Sonic Hedgehog (rhShh). In vivo, itraconazole significantly reduced tumor growth in A375 and A2058 xenograft models. Conclusion Itraconazole induces autophagy-mediated apoptosis in melanoma cells by inhibiting Hedgehog signaling, underscoring its potential as a therapeutic option for melanoma treatment.
Collapse
Affiliation(s)
- Shunqiao Jin
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Dermatology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Xiaojiao Liu
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of Dermatology, Chengdu Badachu Medical Aesthetics Hospital, Chengdu, China
| | - Lingqin Cai
- Department of Dermatology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
- Department of Dermatology, Taizhou Rehabilitation Hospital, Taizhou Enze Medical Center (Group), Taizhou, China
| | - Jiayu Yan
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ling Li
- Department of Dermatology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Hongjun Dong
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuxue Gao
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xicong Zhu
- Department of Dermatology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Cong Zhang
- Department of Preventive Medicine, Dalian Medical University, Dalian, China
| | - Xuezhu Xu
- Department of Dermatology, Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
35
|
Chen X, Tian Q, Gao M, Zhou X, Zhou C. Lingguizhugan decoction enhances autophagy of Alzheimer's disease via regulating the mTOR/ p70s6K pathway in vivo and in vitro. Front Aging Neurosci 2025; 17:1478199. [PMID: 39911633 PMCID: PMC11794305 DOI: 10.3389/fnagi.2025.1478199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
Introduction Lingguizhugan decoction (LGZG) has been reported to treat Alzheimer's disease (AD) by anti-inflammatory and transporting amyloid-β (Aβ). Methods Using APP/PS1 transgenic mice as in vivo model and gave LGZG decoction by oral gavage. Using Aβ25-35-induced SH-SY5Y cells as in vitro model and then added LGZG medicated serum (LMS) to observe the regulatory effect of LGZG on AD autophagy-related pathways. Morris water maze (MWM) was used to evaluate the mice's learning and memory ability. Mice's hippocampus tissue sections were stained immunohistochemically to observe hippocampal Aβ deposition. Transmission electron microscopy monitored autophagosomes and autolysosomes. Western blot analysis measured protein expression levels of beclin-1, p62 and light chain 3II (LC3 II) and mTOR signaling. Results: LGZG could greatly improve learning and memory ability of APP/PS1 mice, and enhance autophagy in vitro and in vivo. LGZG increased the levels of beclin-1 and LC3 II and decreased the levels of p62. Conclusion LGZG enhanced autophagy and showed therapeutic potential in AD by inhibiting mTOR/p70s6K signaling.
Collapse
Affiliation(s)
| | | | | | - Xibin Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
36
|
Yifan D, Jiaheng Z, Yili X, Junxia D, Chao T. CircRNA: A new target for ischemic stroke. Gene 2025; 933:148941. [PMID: 39270759 DOI: 10.1016/j.gene.2024.148941] [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: 01/04/2024] [Revised: 05/22/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Ischemic stroke, a clinical emergency and disease with a poor prognosis, has a negative impact on the survival index of patients. It is frequently precipitated by a multitude of risk factors, including trauma. Currently, there is a paucity of predictive indicators for early intervention. As stable and abundant RNA in the body, circRNAs play a regulatory role in miRNAs and proteins, which affect the occurrence and development of diseases. Moreover, circRNAs can serve as predictors of clinical diseases. Several studies have demonstrated that circRNAs play pivotal roles in numerous aspects of ischemic stroke. Consequently, circRNAs have emerged as key areas of investigation in the field of ischemic stroke.
Collapse
Affiliation(s)
- Dong Yifan
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhang Jiaheng
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Xiao Yili
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China
| | - Duan Junxia
- The first affiliated hospital of hunan university of Chinese medicine, Changsha 410007, China
| | - Tan Chao
- Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, China; The first affiliated hospital of hunan university of Chinese medicine, Changsha 410007, China.
| |
Collapse
|
37
|
Zhang J, Qu Z, Xiao X, Adelson DL, Wang F, Wei A, Harata-Lee Y, Cui J, He D, Xie L, Sun L, Li J, Huang Z, Aung T, Yao H, Lin L. A novel sensitizer reduces EGFR-TKI resistance by regulating the PI3K/Akt/mTOR pathway and autophagy. Heliyon 2025; 11:e41104. [PMID: 39844968 PMCID: PMC11750466 DOI: 10.1016/j.heliyon.2024.e41104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Background The incidence and mortality of lung cancer are high, and treatment with epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) is the preferred first-line treatment for patients suffering from non-small cell lung cancer (NSCLC) with EGFR mutations. However, EGFR-TKI resistance leads to treatment failure. Yifei-Sanjie pill (YFSJ) is a novel type of Chinese patent medicine for lung cancer. The development of YFSJ has progressed for more than 30 years; however, little is known about the molecular mechanisms associated with the inhibition of drug resistance. Methods In this study, flow cytometry and transcriptome sequencing were used in vitro to explore the anticancer effect of Yifei-Sanjie pill (YFSJ) on EGFR-TKI-resistant cell lines and to identify potential molecular mechanisms associated with the inhibition of drug resistance. Results We found that in vitro, YFSJ and YFSJ combined with gefitinib significantly reduced the viability of H1975 and H1650 cells, which is dose-dependent at 24 and 48 h. PI3K, Akt and mTOR were downregulated, while after 24 and 48 h of treatment with YFSJ alone and in combination with gefitinib, LC3A and LC3B were up-regulated in both cell lines. Conclusion YFSJ reduced the viability of EGFR-TKI-resistant cell lines, reducing resistance to gefitinib. This might be caused by a decrease in the PI3K/Akt/mTOR pathway and an increase in autophagy.
Collapse
Affiliation(s)
- Jue Zhang
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Zhipeng Qu
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Xi Xiao
- Research Station, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - David L. Adelson
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Funeng Wang
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Aisheng Wei
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Yuka Harata-Lee
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, South Australia, Adelaide, Australia
| | - Jian Cui
- Department of Immunology, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Dongying He
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Le Xie
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Lingling Sun
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Jing Li
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, PR China
| | - Zijing Huang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| | - Thazin Aung
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - Hong Yao
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong Province, PR China
| | - Lizhu Lin
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, PR China
| |
Collapse
|
38
|
Kumar D, Bishnoi M, Kondepudi KK, Sharma SS. Gut Microbiota-Based Interventions for Parkinson's Disease: Neuroprotective Mechanisms and Current Perspective. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10433-x. [PMID: 39809955 DOI: 10.1007/s12602-024-10433-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Recent evidence links gut microbiota alterations to neurodegenerative disorders, including Parkinson's disease (PD). Replenishing the abnormal composition of gut microbiota through gut microbiota-based interventions "prebiotics, probiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT)" has shown beneficial effects in PD. These interventions increase gut metabolites like short-chain fatty acids (SCFAs) and glucagon-like peptide-1 (GLP-1), which may protect dopaminergic neurons via the gut-brain axis. Neuroprotective effects of these interventions are mediated by several mechanisms, including the enhancement of neurotrophin and activation of the PI3K/AKT/mTOR signaling pathway, GLP-1-mediated gut-brain axis signaling, Nrf2/ARE pathway, and autophagy. Other pathways, such as free fatty acid receptor activation, synaptic plasticity improvement, and blood-brain and gut barrier integrity maintenance, also contribute to neuroprotection. Furthermore, the inhibition of the TLR4/NF-кB pathway, MAPK pathway, GSK-3β signaling pathway, miR-155-5p-mediated neuroinflammation, and ferroptosis could account for their protective effects. Clinical studies involving gut microbiota-based interventions have shown therapeutic benefits in PD patients, particularly in improving gastrointestinal dysfunction and some neurological symptoms. However, the effectiveness in alleviating motor symptoms remains mild. Large-scale clinical trials are still needed to confirm these findings. This review emphasizes the neuroprotective mechanisms of gut microbiota-based interventions in PD as supported by both preclinical and clinical studies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
39
|
Liu Y, Wang Y, Yan P, Cui N, Xu K, Liu D, Tian Y, Cao L. NLRP3 Inflammasome-Mediated Osteoarthritis: The Role of Epigenetics. BIOLOGY 2025; 14:71. [PMID: 39857301 PMCID: PMC11761621 DOI: 10.3390/biology14010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
The prevalence of osteoarthritis (OA) notably surges with age and weight gain. The most common clinical therapeutic drugs are painkillers, yet they cannot impede the deteriorating course of OA. Thus, understanding OA's pathogenesis and devising effective therapies is crucial. It is generally recognized that inflammation, pyroptosis, and OA progression are tightly linked. The activation of NLRP3 inflammasome can lead to the discharge of the pro-inflammatory cytokines Interleukin-1β and IL-18, intensifying subsequent inflammatory reactions and promoting OA development. Conversely, the imbalance caused by deacetylase-regulated NLRP3 inflammasome underlies the chronic mild inflammation related to degenerative diseases. Therefore, this article expounds on the mechanism of OA pathogenesis and the role of histone deacetylases (HDACs) in NLRP3 inflammasome-triggered OA, and illustrates the application of HDAC inhibitors in OA, striving to provide more insights into novel OA treatment approaches.
Collapse
Affiliation(s)
- Yuzhou Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Ying Wang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Ping Yan
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (P.Y.); (N.C.)
| | - Ning Cui
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (P.Y.); (N.C.)
| | - Kejin Xu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (Y.L.); (Y.W.); (K.X.)
| | - Da Liu
- Public Laboratory Centre, Changchun University of Chinese Medicine, Changchun 130117, China;
| | - Yuan Tian
- Clinical School of Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China
| | - Lingling Cao
- Clinical School of Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China
| |
Collapse
|
40
|
Hou Y, Wang Z, Wu Y, Deng T, Yang S, Ma S, Zhang Y, Luo D, Chen Y, Dai X. Sophaline B inhibits non-small cell lung cancer by activating NLRP3/caspase-1/GSDMD-dependent pyroptosis and PI3K/AKT/mTOR-mediated autophagy. Nat Prod Res 2025:1-13. [PMID: 39797590 DOI: 10.1080/14786419.2024.2448839] [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: 05/12/2023] [Revised: 12/01/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Sophaline B (SPB), extracted from the seeds of Sophora alopecuroides L., is a natural bioactive compound that effectively exerts antiviral activities against the hepatitis B virus. This is the first study to demonstrate that SPB exerts anti-tumor effects on NSCLC by inducing pyroptosis and autophagy. SPB promotes NSCLC cell death by increasing the reactive oxygen species (ROS) production to induce pyroptosis via activating the NLRP3/Caspase-1/GSDMD signalling pathway. Meanwhile, SPB inhibits cancer cell proliferation by activating autophagy via blocking the PI3K/AKT/mTOR signalling pathway. Further investigation indicates that SPB inhibits NSCLC cell migration and invasion by increasing E-cadherin while decreasing N-cadherin, vimentin, and snail at the protein level. In addition, our results show that SPB inhibits cancer cell colony formation and human umbilical vein endothelial cell angiogenesis. Our study revealed the mechanisms of SPB triggering pyroptosis and autophagy in NSCLC, thus providing evidence and information on the SPB as an anti-cancer agent in NSCLC.
Collapse
Affiliation(s)
- Yibo Hou
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Zhongfu Wang
- Department of Interventional, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Yang Wu
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Ting Deng
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Siyu Yang
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| | - Yubo Zhang
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Ding Luo
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yanjun Chen
- Department of Subject Service and Consultation, Jinan University Library, Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoyong Dai
- Institute of Biopharmaceutical and Health Engineering, State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Gene and Antibody Therapy, Shenzhen International Graduate School, Tsinghua University, Shenzhen, Guangdong, China
| |
Collapse
|
41
|
Zheng Q, Jin X, Nguyen TTM, Yi EJ, Park SJ, Yi GS, Yang SJ, Yi TH. Autophagy-Enhancing Properties of Hedyotis diffusa Extracts in HaCaT Keratinocytes: Potential as an Anti-Photoaging Cosmetic Ingredient. Molecules 2025; 30:261. [PMID: 39860131 PMCID: PMC11767327 DOI: 10.3390/molecules30020261] [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: 11/27/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The decline in autophagy disrupts homeostasis in skin cells, leading to oxidative stress, energy deficiency, and inflammation-all key contributors to skin photoaging. Consequently, activating autophagy has become a focal strategy for delaying skin photoaging. Natural plants are rich in functional molecules and widely used in the development of anti-photoaging cosmetics. Hedyotis diffusa (HD), as a medicinal plant, is renowned for its anti-inflammatory and anticancer properties; however, its effects on skin photoaging remain unclear. This study investigates HD's potential to counteract skin photoaging by restoring mitochondrial autophagy in keratinocytes. We used HPLC to detect the main chemical components in HD and, using a UVB-induced photoaging model in HaCaT keratinocytes, examined the effects of HD on reactive oxygen species (ROS) levels, Ca2+ concentration, mitochondrial membrane potential (MMP), apoptosis, and the cell cycle. Cellular respiration was further evaluated with the Seahorse XFp Analyzer, and RT-PCR and Western blotting were used to analyze the impact of HD on mitochondrial autophagy-related gene expression and signaling pathways. Our findings indicate that HD promotes autophagy by modulating the PI3K/AKT/mTOR and PINK/PARK2 pathways, which stabilizes mitochondrial quality, maintains MMP and Ca2+ balance, and reduces cytochrome c release. These effects relieve cell cycle arrest and prevent apoptosis associated with an increased BAX/BCL-2 ratio. Thus, HD holds promise as an effective anti-photoaging ingredient with potential applications in the development of cosmetic products.
Collapse
Affiliation(s)
- Qiwen Zheng
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| | - Xiangji Jin
- Department of Dermatology, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Trang Thi Minh Nguyen
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| | - Eun-Ji Yi
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| | - Se-Jig Park
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| | - Gyeong-Seon Yi
- Department of Biopharmaceutical Biotechnology, Graduate School, Kyung Hee University, Yongin-si 17104, Republic of Korea;
| | - Su-Jin Yang
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| | - Tae-Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University, Yongin-si 17104, Republic of Korea; (Q.Z.); (T.T.M.N.); (E.-J.Y.); (S.-J.P.); (S.-J.Y.)
| |
Collapse
|
42
|
Gómez Armengol E, Merckx C, De Sutter H, De Bleecker JL, De Paepe B. Changes to the Autophagy-Related Muscle Proteome Following Short-Term Treatment with Ectoine in the Duchenne Muscular Dystrophy Mouse Model mdx. Int J Mol Sci 2025; 26:439. [PMID: 39859157 PMCID: PMC11765399 DOI: 10.3390/ijms26020439] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
The most severe form of muscular dystrophy (MD), known as Duchenne MD (DMD), remains an incurable disease, hence the ongoing efforts to develop supportive therapies. The dysregulation of autophagy, a degradative yet protective mechanism activated when tissues are under severe and prolonged stress, is critically involved in DMD. Treatments that harness autophagic capacities therefore represent a promising therapeutic approach. Osmolytes are protective organic molecules that regulate osmotic pressure and cellular homeostasis and may support tissue-repairing autophagy. We therefore explored the effects of the osmolyte ectoine in the standard mouse model of DMD, the mdx, focusing on the autophagy-related proteome. Mice were treated with ectoine in their drinking water (150 mg/kg) or through daily intraperitoneal injection (177 mg/kg) until they were 5.5 weeks old. Hind limb muscles were dissected, and samples were prepared for Western blotting for protein quantification and for immunofluorescence for an evaluation of tissue distribution. We report changes in the protein levels of autophagy-related 5 (ATG5), Ser366-phosphorylated sequestosome 1 (SQSTM1), heat shock protein 70 (HSP70), activated microtubule-associated protein 1A/1B-light chain 3 (LC3 II) and mammalian target of rapamycin (mTOR). Most importantly, ectoine significantly improved the balance between LC3 II and SQSTM1 levels in mdx gastrocnemius muscle, and LC3 II immunostaining was most pronounced in muscle fibers of the tibialis anterior from treated mdx. These findings lend support for the further investigation of ectoine as a potential therapeutic intervention for DMD.
Collapse
Affiliation(s)
| | | | | | | | - Boel De Paepe
- Neuromuscular Reference Center and Department of Neurology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium (J.L.D.B.)
| |
Collapse
|
43
|
De S, Banerjee S, Rakshit P, Banerjee S, Kumar SKA. Unraveling the Ties: Type 2 Diabetes and Parkinson's Disease - A Nano-Based Targeted Drug Delivery Approach. Curr Diabetes Rev 2025; 21:32-58. [PMID: 38747222 DOI: 10.2174/0115733998291968240429111357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/04/2024] [Accepted: 04/15/2024] [Indexed: 02/26/2025]
Abstract
The link between Type 2 Diabetes (T2DM) and Parkinson's Disease (PD) dates back to the early 1960s, and ongoing research is exploring this association. PD is linked to dysregulation of dopaminergic pathways, neuroinflammation, decreased PPAR-γ coactivator 1-α, increased phosphoprotein enriched in diabetes, and accelerated α-Syn amyloid fibril production caused by T2DM. This study aims to comprehensively evaluate the T2DM-PD association and risk factors for PD in T2DM individuals. The study reviews existing literature using reputable sources like Scopus, ScienceDirect, and PubMed, revealing a significant association between T2DM and worsened PD symptoms. Genetic profiles of T2DM-PD individuals show similarities, and potential risk factors include insulin-resistance and dysbiosis of the gut-brain microbiome. Anti-diabetic drugs exhibit neuroprotective effects in PD, and nanoscale delivery systems like exosomes, micelles, and liposomes show promise in enhancing drug efficacy by crossing the Blood-Brain Barrier (BBB). Brain targeting for PD uses exosomes, micelles, liposomes, dendrimers, solid lipid nanoparticles, nano-sized polymers, and niosomes to improve medication and gene therapy efficacy. Surface modification of nanocarriers with bioactive compounds (such as angiopep, lactoferrin, and OX26) enhances α-Syn conjugation and BBB permeability. Natural exosomes, though limited, hold potential for investigating DM-PD pathways in clinical research. The study delves into the underlying mechanisms of T2DM and PD and explores current therapeutic approaches in the field of nano-based targeted drug delivery. Emphasis is placed on resolved and ongoing issues in understanding and managing both conditions.
Collapse
Affiliation(s)
- Sourav De
- Department of Pharmaceutical Technology, Eminent College of Pharmaceutical Technology, Kolkata, 700126, West Bengal, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - Pallabita Rakshit
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Subhasis Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol, 713301, West Bengal, India
| | - S K Ashok Kumar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore, 632014, Tamil Nadu, India
| |
Collapse
|
44
|
Zhang Z, He T, Gu H, Zhao Y, Tang S, Han K, Hu Y, Wang H, Yu H. Single-cell RNA sequencing identifies the expression of hemoglobin in chondrocyte cell subpopulations in osteoarthritis. BMC Mol Cell Biol 2024; 25:28. [PMID: 39736555 DOI: 10.1186/s12860-024-00519-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/02/2024] [Indexed: 01/01/2025] Open
Abstract
In recent years, chondrocytes have been found to contain hemoglobin, which might be an alternative strategy for adapting to the hypoxic environment, while the potential mechanisms of that is still unclear. Here, we report the expression characteristics and potential associated pathways of hemoglobin in chondrocytes using single-cell RNA sequencing (scRNA-seq). We downloaded data of normal people and patients with osteoarthritis (OA) from the Gene Expression Omnibus (GEO) database and cells are unbiased clustered based on gene expression pattern. We determined the expression levels of hemoglobin in various chondrocyte subpopulations. Meanwhile, we further explored the difference in the enriched signaling pathways and the cell-cell interaction in chondrocytes of the hemoglobin high-expression and low-expression groups. Specifically, we found that SPP1 was closely associated with the expression of hemoglobin in OA progression. Our findings provide new insights into the distribution characteristics of hemoglobin in chondrocytes and provide potential clues to the underlying role of hemoglobin in OA and the mechanisms related to that, providing potential new ideas for the treatment of OA.
Collapse
Affiliation(s)
- Zhihao Zhang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Ting He
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongwen Gu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yuanhang Zhao
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Shilei Tang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Kangen Han
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Yin Hu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China
| | - Hongwei Wang
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| | - Hailong Yu
- General Hospital of Northern Theater Command, Shenyang, Liaoning Province, 110000, China.
| |
Collapse
|
45
|
Wang Q, Liu X, Yuan J, Yang T, Ding L, Song B, Xu Y. Nek6 regulates autophagy through the mTOR signaling pathway to alleviate cerebral ischemia-reperfusion injury. Mol Brain 2024; 17:96. [PMID: 39702325 DOI: 10.1186/s13041-024-01166-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVE Cerebral ischemia-reperfusion injury (CIRI) is a major obstacle to neurological recovery after clinical treatment of ischemic stroke. The aim of this study was to investigate the molecular mechanism of Nek6 alleviating CIRI through autophagy after cerebral ischemia. MATERIALS AND METHODS A mouse model of CIRI was constructed by middle cerebral artery occlusion (MCAO). TUNEL staining was used to observe the apoptosis of neuronal cells. The oxygen glucose deprivation/reoxygenation (OGD/R) model was established by hypoxia and reoxygenation. The cell apoptosis and activity was detected. Western blot was performed to detect the expression of autophagy-related proteins, protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and adenosine 5'-monophosphate-activated protein kinase (AMPK)/mTOR signaling pathway-related proteins. Cellular autophagy flux was observed by fluorometric method. NIMA-related kinase 6 (Nek6) mRNA stability was detected by actinomycin D treatment. Methylation RNA immunoprecipitation technique was used to detect Nek6 methylation level. RESULTS Nek6 expression was increased in both MCAO and OGD/R models. Overexpression of Nek6 in OGD/R inhibited apoptosis, decreased LC3II and Beclin-1 expression, increased p62 expression, and occurred lysosome dysfunction. Interference with Nek6 has opposite results. Nek6 overexpression promoted p-Akt and p-mTOR protein expressions, inhibited p-AMPK and p-UNC-51-like kinase 1 protein expressions and cell apoptosis, while LY294002, Rapamycin or RSVA405 treatment reversed this effect. Abnormal methyltransferase·like protein 3 (METTL3) expression in CIRI enhanced m6A modification and promoted Nek6 expression level. CONCLUSION This study confirmed that Nek6 regulates autophagy and alleviates CIRI through the mTOR signaling pathway, which provides a novel therapeutic strategy for patients with ischemic stroke in the future.
Collapse
Affiliation(s)
- Qingzhi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Xinjing Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Jing Yuan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Ting Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Lan Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Bo Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jian-She Road, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, China.
| |
Collapse
|
46
|
Mao JQ, Cheng L, Zhang YD, Xie GJ, Wang P. Chinese formula Guben-Jiannao Ye alleviates the dysfunction of circadian and sleep rhythms in APP/PS1 mice implicated in activation of the PI3K/AKT/mTOR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118696. [PMID: 39151711 DOI: 10.1016/j.jep.2024.118696] [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: 05/05/2024] [Revised: 06/24/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Chinese formula Guben-Jiannao Ye (GBJNY) formula has a long history of usage in traditional Chinese medicine (TCM) for the treatment of learning and memory disorders as well as senile insomnia. This formulation is derived from Sun Simiao's five tonic pills. Furthermore, modern pharmacological investigations have revealed its ability to improve cognitive impairment and ameliorate sleep-wake circadian rhythm disorders. However, the precise mechanism underlying its efficacy remains elusive. AIM OF THE STUDY The current research explored the modulatory effects and possible mechanisms of GBJNY in circadian rhythm sleep-wake disorders and cognitive dysfunction in Alzheimer's disease using transcriptome sequencing and experimental validation. MATERIALS AND METHODS The LC-MS/MS tandem technology was utilized to qualitatively discern the active components present in GBJNY. The APP/PS1 mice received continuous treatment with GBJNY or Melatonin for 3 months. The learning and memory abilities of mice were assessed utilizing the Morris water maze (MWM) test, while sleep changes were studied utilizing the electroencephalogram (EEG) and electromyogram (EMG). Concurrently, mice's hippocampus clock gene rhythmicity was investigated. Subsequently, we employed HE staining, Golgi staining, and immunofluorescence to observe GBJNY's impact on synaptic damage and neuronal loss. We performed high-throughput sequencing to analyze the mRNA expression profiles of mice, aiming to identify differentially expressed genes (DEGs). Subsequently, we conducted GO and KEGG enrichment analyses to explore associated signaling pathways. Furthermore, we evaluated the expression levels of proteins involved in the PI3K/AKT/mTOR pathway and Aβ deposition in the hippocampus of mice. Through this comprehensive approach, we sought to elucidate and validate the potential mechanisms of action of GBJNY in APP/PS1 mice. RESULTS Results showed 216 DEGs. Following this, we conducted GO enrichment and KEGG pathway analyses to delve deeper into the distinctions and fundamental functions of the mRNA target genes. The enrichment analysis underscored the prominence of the PI3K/Akt/mTOR signaling pathway as the most pivotal among them. Through in vivo experiments, it was further demonstrated that the administration of GBJNY enhanced memory and learning capacities in APP/PS1 mice. Additionally, GBJNY treatment resulted in alterations in the sleep-wake circadian rhythm, characterized by reduced wakefulness and an increase in non-rapid eye movement (NREM) sleep. Moreover, alterations in the peak expression of Per1, Per2, Clock, Cry1, Cry2, and Bmal1 mRNA were noted in the hippocampus of treated mice. Particularly noteworthy were the observed reductions in amyloid-beta (Aβ) deposition within the hippocampus, improvements in neuronal synaptic integrity, and upregulation of mTOR, Akt, and PI3K protein expression in the hippocampal region. These findings underscore the critical involvement of the PI3K/Akt/mTOR signaling pathway in mitigating disturbances in sleep-wake circadian rhythms. CONCLUSIONS GBJNY enhanced the cognitive performance of APP/PS1 mice and altered clock gene expression patterns, alleviating sleep-wake circadian rhythm disruptions. The fundamental mechanism appears to be linked to the PI3K/Akt/mTOR pathway regulation, offering a foundation for potential clinical applications.
Collapse
Affiliation(s)
- Jian-Qin Mao
- Basic Medicine College, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Li Cheng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Yu-Dan Zhang
- Health Medical Center, Hubei Minzu University, Enshi, 445000, China.
| | - Guang-Jing Xie
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Ping Wang
- Engineering Research Center, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
47
|
Lee SH, Bae EJ, Perez-Acuna D, Jung MK, Han JW, Mook-Jung I, Lee SJ. Amyloid-β-activated microglia can induce compound proteinopathies. Brain 2024; 147:4105-4120. [PMID: 39194073 DOI: 10.1093/brain/awae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 08/29/2024] Open
Abstract
Neuropathological features of Alzheimer's disease include amyloid plaques, neurofibrillary tangles and Lewy bodies, with the former preceding the latter two. However, it is not fully understood how these compound proteinopathies are interconnected. Here, we show that transplantation of amyloid-β oligomer-activated microglia into the striatum of naïve mice was sufficient to generate all the features of Alzheimer's disease, including widespread tauopathy and synucleinopathy, gliosis, neuroinflammation, synapse loss, neuronal death, and cognitive and motor deficits. These pathological features were eliminated by microglia depletion and anti-inflammatory drug administration. Our results suggest the crucial roles of microglia-driven inflammation in development of mixed pathology. This study provides not only mechanistic insights into amyloid-β oligomer-triggered proteinopathies but also a novel animal model recapitulating the salient features of Alzheimer's disease.
Collapse
Affiliation(s)
- Sang Hwan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Eun-Jin Bae
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Dayana Perez-Acuna
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Min Kyo Jung
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu 41068, Korea
| | - Jong Won Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biochemistry, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
- Department of Biochemistry, Seoul National University College of Medicine, Seoul 03080, Korea
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
- Convergence Research Center for Dementia, Medical Research Center, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
48
|
Liu H, Wang J, Yue G, Xu J. Placenta-derived mesenchymal stem cells protect against diabetic kidney disease by upregulating autophagy-mediated SIRT1/FOXO1 pathway. Ren Fail 2024; 46:2303396. [PMID: 38234193 PMCID: PMC10798286 DOI: 10.1080/0886022x.2024.2303396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
Diabetic kidney disease (DKD) is a common chronic microvascular complication of diabetes mellitus. Although studies have indicated the therapeutic potential of mesenchymal stem cells (MSCs) for DKD, the underlying molecular mechanisms remain unclear. Herein, we explored the renoprotective effect of placenta-derived MSCs (P-MSCs) and the potential mechanism of SIRT1/FOXO1 pathway-mediated autophagy in DKD. The urine microalbumin/creatinine ratio was determined using ELISA, and renal pathological changes were detected by special staining techniques. Immunofluorescence was used for detecting the renal tissue expression of podocin and nephrin; immunohistochemistry for the renal expression of autophagy-related proteins (LC3, Beclin-1, SIRT1, and FOXO1); and western blotting and PCR for the expression of podocyte autophagy- and pathway-related indicators. We found that P-MSCs ameliorated renal tubular injury and glomerular mesangial matrix deposition and alleviated podocyte damage in DKD rats. PMSCs enhanced autophagy levels and increased SIRT1 and FOXO1 expression in DKD rat renal tissue, whereas the autophagy inhibitor 3-methyladenine significantly attenuated the renoprotective effect of P-MSCs. P-MSCs improved HG-induced Mouse podocyte clone5(MPC5)injury, increased podocyte autophagy, and upregulated SIRT1 and FOXO1 expression. Moreover, downregulation of SIRT1 expression blocked the P-MSC-mediated enhancement of podocyte autophagy and improvement of podocyte injury. Thus, P-MSCs can significantly improve renal damage and reduce podocyte injury in DKD rats by modulating the SIRT1/FOXO1 pathway and enhancing podocyte autophagy.
Collapse
Affiliation(s)
- Honghong Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, P.R.China
| | - Jiao Wang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, P.R.China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, P.R.China
- Jiangxi branch of national clinical research center for metabolic disease, Nanchang, P.R.China
| | - Guanru Yue
- Department of Medical Genetics and Cell biology, Medical College of Nanchang University, Nanchang, P.R. China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, P.R.China
- Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, Nanchang, P.R.China
- Jiangxi branch of national clinical research center for metabolic disease, Nanchang, P.R.China
| |
Collapse
|
49
|
Luo S, Huang X, Li S, Chen Y, Zhang X, Zeng X. Homogeneous Polyporus polysaccharide exerts anti-bladder cancer effects via autophagy induction. PHARMACEUTICAL BIOLOGY 2024; 62:214-221. [PMID: 38353262 PMCID: PMC10868468 DOI: 10.1080/13880209.2024.2316195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024]
Abstract
CONTEXT Polyporus polysaccharide (PPS), the leading bioactive ingredient extracted from Polyporus umbellatus (Pers.) Fr. (Polyporaceae), has been demonstrated to exert anti-bladder cancer and immunomodulatory functions in macrophages. OBJECTIVE To explore the effects of homogeneous Polyporus polysaccharide (HPP) on the proliferation and autophagy of bladder cancer cells co-cultured with macrophages. MATERIALS AND METHODS MB49 bladder cancer cells and RAW264.7 macrophages were co-cultured with or without HPP intervention (50, 100, or 200 μg/mL) for 24 h. The cell counting kit-8 (CCK-8) assay and 5-ethynyl-2″-deoxyuridine (EdU) staining evaluated MB49 cell proliferation. Monodansylcadaverine (MDC) staining and transmission electron microscopy (TEM) observed autophagosomes. Western blotting detected the expression levels of autophagy-related proteins and PI3K/Akt/mTOR pathway proteins. RESULTS HPP inhibited the proliferation of MB49 cells co-cultured with RAW264.7 cells but not MB49 cells alone. HPP altered the expression of autophagy-related proteins and promoted the formation of autophagosomes in MB49 cells in the co-culture system. Autophagy inhibitors 3-methyladenine (3-MA) and chloroquine (CQ) not only antagonized HPP-induced autophagy but also attenuated the inhibitory effects of HPP on MB49 cell proliferation in the co-culture system. HPP or RAW264.7 alone was not sufficient to induce autophagy in MB49 cells. In addition, HPP suppressed the protein expression of the PI3K/Akt/mTOR pathway in MB49 cells in the co-culture system. DISCUSSION AND CONCLUSIONS HPP induced bladder cancer cell autophagy by regulating macrophages in the co-culture system, resulting in the inhibition of cancer cell proliferation. The PI3K/Akt/mTOR pathway was involved in HPP-induced autophagy in the co-culture system.
Collapse
Affiliation(s)
- Siwan Luo
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaopeng Huang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shiqi Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuwen Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xian Zhang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xing Zeng
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
50
|
Chen C, Liao J, Sun X. Keratinocyte differentiation factor 1 enhances cervical cancer cell viability and migration by activating the PI3K/AKT pathway. J OBSTET GYNAECOL 2024; 44:2362420. [PMID: 38864525 DOI: 10.1080/01443615.2024.2362420] [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: 09/08/2023] [Accepted: 05/26/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND The aim of This study is to investigate the effects of Keratinocyte differentiation factor 1 (KDF1) on cervical cancer cells and the underlying mechanisms. METHODS The Gene Expression Profiling Interactive Analysis database was used to analyse KDF1 expression in cervical cancer and paracancerous tissue samples. The correlation between the expression of KDF1 and clinicopathological features was also analysed. Cervical cancer cells (HeLa cells) with KDF1 overexpression or knockdown were constructed. Reverse transcription polymerase chain reaction was used to detect the mRNA expression of KDF1 in cervical cancer tissues and cells. In different treatment groups of cervical cancer cells, protein expression of KDF1, cell viability, invasion, and migration were subsequently confirmed by western blotting, CCK-8 assay, transwell assay, and wound healing assay, respectively. A PI3K inhibitor (LY294002) was used to detect the effect of KDF1 on the phosphoinositide 3-kinase (PI3K)/Protein Kinase B (AKT) pathway. RESULTS KDF1 was highly expressed in cervical cancer tissues and cell lines (p < 0.01), and was significantly associated with poor prognosis (p < 0.05). Knockdown of KDF1 in HeLa cells resulted in a significant decrease in cell proliferation, migration, and invasion, as well as phosphorylated PI3K (P-PI3K) and p-AKT levels (p < 0.01). However, KDF1 overexpression activated the PI3K/AKT pathway and significantly enhanced the malignant biological behaviour of cervical cancer cells (p < 0.01). Additionally, the PI3K inhibitor reduced the proliferation, invasion, and migration of HeLa cells overexpressing KDF1 (p < 0.01). CONCLUSION KDF1 enhances cervical cancer viability and migration by activating the PI3K/AKT pathway, and may serve as a therapeutic target for patients with cervical cancer.
Collapse
Affiliation(s)
- Chao Chen
- Department of Obstetrics and Gynecology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | | | - Xingxing Sun
- Department of Obstetrics and Gynecology, Wujin Hospital Affiliated with Jiangsu University, The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| |
Collapse
|