1
|
Meng R, Li Y, Yang X, Cheng Y, Xu M, Zhou L, Wu C, Yu S, Huang W, Wang T, Zhang Q. Polyphenol Mediated Assembly: Tailored Nano-Dredger Unblocks Axonal Autophagosomes Retrograde Transport Traffic Jam for Accelerated Alzheimer's Waste Clearance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2413614. [PMID: 39686827 DOI: 10.1002/adma.202413614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/01/2024] [Indexed: 12/18/2024]
Abstract
Clear-cut evidence has linked defective autophagy to Alzheimer's disease (AD). Recent studies underscore a unique hurdle in AD neuronal autophagy: impaired retrograde axonal transport of autophagosomes, potent enough to induce autophagic stress and neurodegeneration. Nonetheless, pertinent therapy is unavailable. Here, a novel combinational therapy composed of siROCK2 and lithospermic acid B (LA) is introduced, tailored to dredge blocked axonal autophagy by multi-mitigating microtubule disruption, ATP depletion, oxidative stress, and autophagy initiation impediments in AD. Leveraging the recent discovery of multi-interactions between polyphenol LA and siRNA, ε-Poly-L-lysine, and anionic lipid nanovacuoles, LA and siROCK2 are successfully co-loaded into a fresh nano-drug delivery system, LIP@PL-LA/siRC, via a ratio-flexible and straightforward fabrication process. Further modification with the TPL peptide onto LIP@PL-LA/siRC creates a brain-neuron targeted, biocompatible, and pluripotent nanomedicine, named "Nano-dredger" (T-LIP@PL-LA/siRC). Nano-dredger efficiently accelerates axonal retrograde transport and lysosomal degradation of autophagosomes, thereby facilitating the clearance of neurotoxic proteins, improving neuronal complexity, and alleviating memory defects in 3×Tg-AD transgenic mice. This study provides a fresh and flexible polyphenol/siRNA co-delivery paradigm and furnishes conceptual proof that dredging axonal autophagy represents a promising AD therapeutic avenue.
Collapse
Affiliation(s)
- Ran Meng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Yunlong Cheng
- Shanxi Academy of Traditional Chinese Medicine, Xi'an, 710003, P. R. China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - LingLing Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Chengqin Wu
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Shuai Yu
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Wenyi Huang
- Guangzhou CSR Biotech Co. Ltd, Guangzhou, 510700, P. R. China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, School of Pharmacy, Fudan University, Shanghai, 201203, P. R. China
| |
Collapse
|
2
|
Ye Q, Li X, Gao W, Gao J, Zheng L, Zhang M, Yang F, Li H. Role of Rho-associated kinases and their inhibitor fasudil in neurodegenerative diseases. Front Neurosci 2024; 18:1481983. [PMID: 39628659 PMCID: PMC11613983 DOI: 10.3389/fnins.2024.1481983] [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: 08/17/2024] [Accepted: 11/01/2024] [Indexed: 12/06/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are prevalent in the elderly. The pathogenesis of NDDs is complex, and currently, there is no cure available. With the increase in aging population, over 20 million people are affected by common NDDs alone (Alzheimer's disease and Parkinson's disease). Therefore, NDDs have profound negative impacts on patients, their families, and society, making them a major global health concern. Rho-associated kinases (ROCKs) belong to the serine/threonine protein kinases family, which modulate diverse cellular processes (e.g., apoptosis). ROCKs may elevate the risk of various NDDs (including Huntington's disease, Parkinson's disease, and Alzheimer's disease) by disrupting synaptic plasticity and promoting inflammatory responses. Therefore, ROCK inhibitors have been regarded as ideal therapies for NDDs in recent years. Fasudil, one of the classic ROCK inhibitor, is a potential drug for treating NDDs, as it repairs nerve damage and promotes axonal regeneration. Thus, the current review summarizes the relationship between ROCKs and NDDs and the mechanism by which fasudil inhibits ROCKs to provide new ideas for the treatment of NDDs.
Collapse
Affiliation(s)
- Qiuyan Ye
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xue Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- Jiangsu College of Nursing, Huaian, China
| | - Jiayue Gao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liping Zheng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Miaomiao Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fengge Yang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Honglin Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
3
|
Hossen F, Geng X, Sun GY, Yao X, Lee JC. Oligomeric Amyloid-β and Tau Alter Cell Adhesion Properties and Induce Inflammatory Responses in Cerebral Endothelial Cells Through the RhoA/ROCK Pathway. Mol Neurobiol 2024; 61:8759-8776. [PMID: 38561558 PMCID: PMC11445398 DOI: 10.1007/s12035-024-04138-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Dysfunction of cerebral endothelial cells (CECs) has been implicated in the pathology of Alzheimer's disease (AD). Despite evidence showing cytotoxic effects of oligomeric amyloid-β (oAβ) and Tau (oTau) in the central nervous system, their direct effects on CECs have not been fully investigated. In this study, we examined the direct effects of oAβ, oTau, and their combination on cell adhesion properties and inflammatory responses in CECs. We found that both oAβ and oTau increased cell stiffness, as well as the p-selectin/Sialyl-LewisX (sLeX) bonding-mediated membrane tether force and probability of adhesion in CECs. Consistent with these biomechanical alterations, treatments with oAβ or oTau also increased actin polymerization and the expression of p-selectin at the cell surface. These toxic oligomeric peptides also triggered inflammatory responses, including upregulations of p-NF-kB p65, IL-1β, and TNF-α. In addition, they rapidly activated the RhoA/ROCK pathway. These biochemical and biomechanical changes were further enhanced by the treatment with the combination of oAβ and oTau, which were significantly suppressed by Fasudil, a specific inhibitor for the RhoA/ROCK pathway. In conclusion, our data suggest that oAβ, oTau, and their combination triggered subcellular mechanical alterations and inflammatory responses in CECs through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xue Geng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Xincheng Yao
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
4
|
Wang W, Rui M. Advances in understanding the roles of actin scaffolding and membrane trafficking in dendrite development. J Genet Genomics 2024; 51:1151-1161. [PMID: 38925347 DOI: 10.1016/j.jgg.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Dendritic morphology is typically highly branched, and the branching and synaptic abundance of dendrites can enhance the receptive range of neurons and the diversity of information received, thus providing the basis for information processing in the nervous system. Once dendritic development is aberrantly compromised or damaged, it may lead to abnormal connectivity of the neural network, affecting the function and stability of the nervous system and ultimately triggering a series of neurological disorders. Research on the regulation of dendritic developmental processes has flourished, and much progress is now being made in its regulatory mechanisms. Noteworthily, dendrites are characterized by an extremely complex dendritic arborization that cannot be attributed to individual protein functions alone, requiring a systematic analysis of the intrinsic and extrinsic signals and the coordinated roles among them. Actin cytoskeleton organization and membrane vesicle trafficking are required during dendrite development, with actin providing tracks for vesicles and vesicle trafficking in turn providing material for actin assembly. In this review, we focus on these two basic biological processes and discuss the molecular mechanisms and their synergistic effects underlying the morphogenesis of neuronal dendrites. We also offer insights and discuss strategies for the potential preventive and therapeutic treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wanting Wang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China
| | - Menglong Rui
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu 210031, China.
| |
Collapse
|
5
|
Rosewood TJ, Nho K, Risacher SL, Liu S, Gao S, Shen L, Foroud T, Saykin AJ. Pathway enrichment in genome-wide analysis of longitudinal Alzheimer's disease biomarker endophenotypes. Alzheimers Dement 2024. [PMID: 39440837 DOI: 10.1002/alz.14308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION The genetic pathways that influence longitudinal heterogeneous changes in Alzheimer's disease (AD) may provide insight into disease mechanisms and potential therapeutic targets. METHODS Longitudinal endophenotypes from the Alzheimer's Disease Neuroimaging Initiative (ADNI) representing amyloid, tau, neurodegeneration (A/T/N), and cognition were selected. Genome-wide association analysis was performed using a linear mixed model (LMM) approach, followed by gene and pathway enrichment with significant and functionally relevant SNPs. RESULTS A total of 33 and 19 statistically significant pathways were identified associating with the intercept and longitudinal trajectory, respectively. The longitudinal intercept pathways represent eight groups: immune, metabolic, cell growth and survival, DNA maintenance, neuronal signaling, RAS/MAPK/ERK signaling pathways, vesicle and lysosomal transport, and transcription modification. Longitudinal trajectory pathways represented six groups: Immune, metabolic, cell signaling, cytoskeleton, and glycosylation. DISCUSSION Longitudinal enrichment identified pathways that uniquely associate with trajectories of key AD biomarkers and cognition, providing new insight into AD course-related mechanisms and potential new therapeutic targets. HIGHLIGHTS A systematic genome-wide analysis with longitudinal AD biomarker endophenotypes was performed. Enriched pathways were identified with functionally derived SNP to gene analysis. Fifty-two pathways were associated with longitudinal trajectory and intercept. Many of the identified pathways are specific steps in larger pathways implicated in AD. The identified pathways may provide therapeutic targets and areas for further study.
Collapse
Affiliation(s)
- Thea J Rosewood
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- School of Informatics and Computing, Indiana University, Indianapolis, Indiana, USA
| | - Shannon L Risacher
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Shiwei Liu
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tatiana Foroud
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Zhang C, Zheng J, Yu X, Kuang B, Dai X, Zheng L, Yu W, Teng W, Cao H, Li M, Yao J, Liu X, Zou W. "Baihui" (DU20)-penetrating "Qubin" (GB7) acupuncture on blood-brain barrier integrity in rat intracerebral hemorrhage models via the RhoA/ROCK II/MLC 2 signaling pathway. Animal Model Exp Med 2024; 7:740-757. [PMID: 38379356 PMCID: PMC11528382 DOI: 10.1002/ame2.12374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/21/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Blocking the RhoA/ROCK II/MLC 2 (Ras homolog gene family member A/Rho kinase II/myosin light chain 2) signaling pathway can initiate neuroprotective mechanisms against neurological diseases such as stroke, cerebral ischemia, and subarachnoid hemorrhage. Nevertheless, it is not clear whether and how disrupting the RhoA/ROCK II/MLC 2 signaling pathway changes the pathogenic processes of the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH). The present investigation included the injection of rat caudal vein blood into the basal ganglia area to replicate the pathophysiological conditions caused by ICH. METHODS Scalp acupuncture (SA) therapy was performed on rats with ICH at the acupuncture point "Baihui"-penetrating "Qubin," and the ROCK selective inhibitor fasudil was used as a positive control to evaluate the inhibitory effect of acupuncture on the RhoA/ROCK II/MLC 2 signaling pathway. Post-assessments included neurological deficits, brain edema, Evans blue extravasation, Western blot, quantitative polymerase chain reaction, and transmission electron microscope imaging. RESULTS We found that ROCK II acts as a promoter of the RhoA/ROCK II/MLC 2 signaling pathway, and its expression increased at 6 h after ICH, peaked at 3 days, and then decreased at 7 days after ICH, but was still higher than the pre-intervention level. According to some experimental results, although 3 days is the peak, 7 days is the best time point for acupuncture treatment. Starting from 6 h after ICH, the neurovascular structure and endothelial cell morphology around the hematoma began to change. Based on the changes in the promoter ROCK II, a 7-day time point was selected as the breakthrough point for treating ICH model rats in the main experiment. The results of this experiment showed that both SA at "Baihui"-penetrating "Qubin" and treatment with fasudil could improve the expression of endothelial-related proteins by inhibiting the RhoA/ROCK II/MLC 2 signaling pathway and reduce neurological dysfunction, brain edema, and BBB permeability in rats. CONCLUSION This study found that these experimental data indicated that SA at "Baihui"-penetrating "Qubin" could preserve BBB integrity and neurological function recovery after ICH by inhibiting RhoA/ROCK II/MLC 2 signaling pathway activation and by regulating endothelial cell-related proteins.
Collapse
Affiliation(s)
- Ce Zhang
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Jia Zheng
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Xueping Yu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Binglin Kuang
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Xiaohong Dai
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Lei Zheng
- Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine of Heilongjiang University of Chinese MedicineHarbinChina
| | - Weiwei Yu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Wei Teng
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Hongtao Cao
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Mingyue Li
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Jiayong Yao
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Xiaoying Liu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Wei Zou
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
7
|
Butt TH, Tobiume M, Re DB, Kariya S. Physical Exercise Counteracts Aging-Associated White Matter Demyelination Causing Cognitive Decline. Aging Dis 2024; 15:2136-2148. [PMID: 38377028 PMCID: PMC11346408 DOI: 10.14336/ad.2024.0216] [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: 01/14/2024] [Accepted: 02/16/2024] [Indexed: 02/22/2024] Open
Abstract
In the central nervous system, oligodendrocytes wrap around neuronal axons to form myelin, an insulating layer or sheath that allows for the efficient conductance of action potentials. In addition to structural insulation, myelin provides encased axons with nutrient, metabolic and defensive support. Demyelination, or myelin loss, can therefore cause axonal dysfunction, leading to neurological impairment and disease. In Alzheimer's disease (AD), progressive white matter demyelination is acknowledged as one of the earliest pathologies preceding symptom onset. Unfortunately, current pharmacotherapy for slowing demyelination or promoting remyelination in AD is nonexistent. Exercise is recognized for its wide-ranging benefits to human health, including improved mental health and the prevention of lifestyle-related diseases. Mounting evidence suggests the contribution of physical activity in delaying the progression of dementia in elderly populations. Recent mechanistic studies have shown that exercise facilitates myelination in the brain through the vitalization of intrinsic pro-myelination cues, such as increased neurotrophic factors and electrical activity. In this review, we summarize and discuss the potential of physical exercise on counteracting aging-associated white matter demyelination, which causes cognitive decline in AD. We highlight the need of further basic and clinical research investigations on this topic to establish novel approaches for healthy and improved brain aging.
Collapse
Affiliation(s)
- Tanya H Butt
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Makoto Tobiume
- Unit for Respiratory System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| | - Diane B Re
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
- NIEHS Center for Environmental Health Sciences in Northern Manhattan, Columbia University, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
| | - Shingo Kariya
- Unit for Nervous System & Dementia in the Division of Internal Medicine, Katsuren Hospital, Itoman, Okinawa, Japan
| |
Collapse
|
8
|
Hossen F, Sun GY, Lee JC. Oligomeric Tau-induced oxidative damage and functional alterations in cerebral endothelial cells: Role of RhoA/ROCK signaling pathway. Free Radic Biol Med 2024; 221:261-272. [PMID: 38815773 PMCID: PMC11184584 DOI: 10.1016/j.freeradbiomed.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Despite of yet unknown mechanism, microvascular deposition of oligomeric Tau (oTau) has been implicated in alteration of the Blood-Brain Barrier (BBB) function in Alzheimer's disease (AD) brains. In this study, we employed an in vitro BBB model using primary mouse cerebral endothelial cells (CECs) to investigate the mechanism underlying the effects of oTau on BBB function. We found that exposing CECs to oTau induced oxidative stress through NADPH oxidase, increased oxidative damage to proteins, decreased proteasome activity, and expressions of tight junction (TJ) proteins including occludin, zonula occludens-1 (ZO-1) and claudin-5. These effects were suppressed by the pretreatment with Fasudil, a RhoA/ROCK signaling inhibitor. Consistent with the biochemical alterations, we found that exposing the basolateral side of CECs to oTau in the BBB model disrupted the integrity of the BBB, as indicated by an increase in FITC-dextran transport across the model, and a decrease in trans endothelial electrical resistance (TEER). oTau also increased the transmigration of peripheral blood mononuclear cells (PBMCs) in the BBB model. These functional alterations in the BBB induced by oTau were also suppressed by Fasudil. Taken together, our findings suggest that targeting the RhoA/ROCK pathway can be a potential therapeutic strategy to maintain BBB function in AD.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
9
|
Li X, Du H, Zhou H, Huang Y, Tang S, Yu C, Guo Y, Luo W, Gong Y. FOXL2 regulates RhoA expression to change actin cytoskeleton rearrangement in granulosa cells of chicken pre-ovulatory follicles†. Biol Reprod 2024; 111:391-405. [PMID: 38832713 DOI: 10.1093/biolre/ioae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/04/2024] [Accepted: 06/01/2024] [Indexed: 06/05/2024] Open
Abstract
Forkhead box L2 (FOXL2) is an indispensable key regulator of female follicular development, and it plays important roles in the morphogenesis, proliferation, and differentiation of follicle granulosa cells, such as establishing normal estradiol signaling and regulating steroid hormone synthesis. Nevertheless, the effects of FOXL2 on granulosa cell morphology and the underlying mechanism remain unknown. Using FOXL2 ChIP-seq analysis, we found that FOXL2 target genes were significantly enriched in the actin cytoskeleton-related pathways. We confirmed that FOXL2 inhibited the expression of RhoA, a key gene for actin cytoskeleton rearrangement, by binding to TCATCCATCTCT in RhoA promoter region. In addition, FOXL2 overexpression in granulosa cells induced the depolymerization of F-actin and disordered the actin filaments, resulting in a slowdown in the expansion of granulosa cells, while FOXL2 silencing inhibited F-actin depolymerization and stabilized the actin filaments, thereby accelerating granulosa cell expansion. RhoA/ROCK pathway inhibitor Y-27632 exhibited similar effects to FOXL2 overexpression, even reversed the actin polymerization in FOXL2 silencing granulosa cells. This study revealed for the first time that FOXL2 regulated granulosa cell actin cytoskeleton by RhoA/ROCK pathway, thus affecting granulosa cell expansion. Our findings provide new insights for constructing the regulatory network of FOXL2 and propose a potential mechanism for facilitating rapid follicle expansion, thereby laying a foundation for further understanding follicular development.
Collapse
Affiliation(s)
- Xuelian Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Hongting Du
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Haobo Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Ying Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Shuixin Tang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Chengzhi Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yan Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Wei Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Yanzhang Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Wuhan, Hubei, PR China
- College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| |
Collapse
|
10
|
Cai Y, Wang LW, Wu J, Chen ZW, Yu XF, Liu FH, Gao DP. Fasudil alleviates alcohol-induced cognitive deficits and hippocampal morphology injury partly by altering the assembly of the actin cytoskeleton and microtubules. Behav Brain Res 2024; 471:115068. [PMID: 38830386 DOI: 10.1016/j.bbr.2024.115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/07/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024]
Abstract
Alcohol-Related Brain Damage (ARBD) manifests predominantly as cognitive impairment and brain atrophy with the hippocampus showing particular vulnerability. Fasudil, a Rho kinase (ROCK) inhibitor, has established neuroprotective properties; however, its impact on alcohol-induced cognitive dysfunction and hippocampal structural damage remains unelucidated. This study probes Fasudil's neuroprotective potential and identifies its mechanism of action in an in vivo context. Male C57BL/6 J mice were exposed to 30% (v/v, 6.0 g/kg) ethanol by intragastric administration for four weeks. Concurrently, these mice received a co-treatment with Fasudil through intraperitoneal injections at a dosage of 10 mg/kg/day. Fasudil was found to mitigate alcohol-induced spatial and recognition memory deficits, which were quantified using Y maze, Morris water maze, and novel object recognition tests. Concurrently, Fasudil attenuated hippocampal structural damage prompted by chronic alcohol exposure. Notably, Fasudil moderated alcohol-induced disassembly of the actin cytoskeleton and microtubules-mechanisms central to the maintenance of hippocampal synaptic integrity. Collectively, our findings indicate that Fasudil partially reverses alcohol-induced cognitive and morphological detriments by modulating cytoskeletal dynamics, offering insights into potential therapeutic strategies for ARBD.
Collapse
Affiliation(s)
- Yu Cai
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Lu-Wan Wang
- School of Medical, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, PR China
| | - Jing Wu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Zi-Wei Chen
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Xue-Feng Yu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Fu-He Liu
- Department of Pharmacy, Zhejiang Pharmaceutical University, 666 Siming Rd, Ningbo, Zhejiang 315500, PR China
| | - Da-Peng Gao
- Department of Neurology, The First Affiliated Hospital of Ningbo University, 247 Renmin Rd, Ningbo, Zhejiang 315020, PR China.
| |
Collapse
|
11
|
Ke C, Shan S, Tan Y, Cao Y, Xie Z, Shi S, Pan J, Zhang W. Signaling pathways in the treatment of Alzheimer's disease with acupuncture: a narrative review. Acupunct Med 2024; 42:216-230. [PMID: 38859546 DOI: 10.1177/09645284241256669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
BACKGROUND To date, there is no effective treatment for Alzheimer's disease (AD), a progressive neurodegenerative disorder that is increasing in prevalence worldwide. The objective of this review was to summarize the core targets and signaling pathways involved in acupuncture treatment for AD. METHODS We reviewed numerous signaling pathways, including mammalian target of rapamycin (mTOR), phosphatidylinositol 3-kinase-protein kinase B (PI3 K/Akt), adenosine monophosphate-activated protein kinase (AMPK), mitogen-activated protein kinase (MAPK), nuclear factor (NF)-kB, p53, Wnt, nitric oxide (NO), Janus kinase / signal transducer and activator of transcription (JAK/ STAT), RhoA/ROCK (Rho-associated protein kinase) and Ca2+/ calmodulin-dependent protein kinase II (CaMKII) / cyclic adenosine monophosphate-response element-binding protein (CREB). The relevant data were obtained from PubMed, EMBASE, Web of Science, China National Knowledge Infrastructure (CNKI) and Wanfang databases. RESULTS In summary, the effects of acupuncture are mediated by multiple targets and pathways. Furthermore, acupuncture can improve pathological changes associated with AD (such as abnormal deposition of amyloid (A)β, tau hyperphosphorylation, synaptic dysfunction and neuronal apoptosis) through multiple signaling pathways. CONCLUSION Overall, our findings provide a basis for future research into the effects of acupuncture on AD.
Collapse
Affiliation(s)
- Chao Ke
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shengtao Shan
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yan Tan
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yang Cao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Zhengrong Xie
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Senjie Shi
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jiang Pan
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Wei Zhang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
12
|
Hu Q, Zhang X, Peng H, Guan J, Huang Z, Jiang B, Sun D. A New Modulator of Neuroinflammation in Diabetic Retinopathy: USP25. Inflammation 2024; 47:1520-1535. [PMID: 38436811 PMCID: PMC11343827 DOI: 10.1007/s10753-024-01991-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/05/2024]
Abstract
Diabetic retinopathy (DR) is a diabetes-associated complication that poses a threat to vision, distinguished by persistent and mild inflammation of the retinal microvasculature. The activation of microglia plays a crucial role in driving this pathological progression. Previous investigations have demonstrated that ubiquitin-specific peptidase 25 (USP25), a deubiquitinating enzyme, is involved in the regulation of immune cell activity. Nevertheless, the precise mechanisms through which USP25 contributes to the development of DR remain incompletely elucidated. Firstly, we have demonstrated the potential mechanism by which ROCKs can facilitate microglial activation and augment the synthesis of inflammatory mediators through the modulation of NF-κB signaling pathways in a high-glucose milieu. Furthermore, our study has provided novel insights by demonstrating that the regulatory role of USP25 in the secretion of proinflammatory factors is mediated through the involvement of ROCK in modulating the expression of NF-κB and facilitating the nuclear translocation of the phosphatase NF-κB. This regulatory mechanism plays a crucial role in modulating the activation of microglial cells within a high-glycemic environment. Hence, USP25 emerges as a pivotal determinant for the inflammatory activation of microglial cells, and its inhibition exhibits a dual effect of promoting retinal neuron survival while suppressing the inflammatory response in the retina. In conclusion, the promotion of diabetic retinopathy (DR) progression by USP25 is attributed to its facilitation of microglial activation induced by high glucose levels, a process mediated by the ROCK pathway. These findings highlight the importance of considering USP25 as a potential therapeutic target for the management of diabetic neuroinflammation.
Collapse
Affiliation(s)
- Qiang Hu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongsong Peng
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jitian Guan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhangxin Huang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Jiang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China
| | - Dawei Sun
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, 157 Baojian Road, Harbin, 150086, China.
| |
Collapse
|
13
|
Yang J, Xiao S, Li L, Zhu A, Xiao W, Wang Q. Actin Dysregulation Mediates Nephrotoxicity of Cassiae Semen Aqueous Extracts. TOXICS 2024; 12:556. [PMID: 39195658 PMCID: PMC11360101 DOI: 10.3390/toxics12080556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024]
Abstract
Cassiae semen, commonly consumed as roasted tea, has been widely used for both medicinal purposes and dietary supplements. In this study, we investigated the nephrotoxic effects and underlying mechanisms of Cassiae semen aqueous extracts (CSAEs) using computational and animal models. Both male and female Sprague Dawley rats were treated with 4.73-47.30 g/kg (body weight) of CSAEs by oral gavage twice a day for 7-28 days. We found that serum and urinary biomarkers of kidney injury and kidney coefficients were increased in a dose-dependent manner, and were accompanied by morphological alterations in the kidneys of CSAEs-treated rats. Computational and molecular docking approaches predicted that the three most abundant components of CSAEs-obtusifolin, aurantio-obtusin, and obtusin-exhibited strong affinity for the binding of F-actin, ROCK1, and Rac1, and the RhoA-ROCK pathway was identified as the most likely regulatory mechanism mediating the nephrotoxicity of CSAEs. Consistently, immunofluorescence staining revealed F-actin and cytoskeleton were frequently disturbed in renal cells and brush borders at high doses of CSAEs. Results from gene expression analyses confirmed that CSAEs suppressed the key proteins in the RhoA-ROCK signaling pathway and consequently the expression of F-actin and its stabilization genes. In summary, our findings suggest that Cassiae semen can depolymerize and destabilize actin cytoskeleton by inhibition of the RhoA-ROCK pathway and/or direct binding to F-actin, leading to nephrotoxicity. The consumption of Cassiae semen as a supplement and medicine warrants attention.
Collapse
Affiliation(s)
- Jinlan Yang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Sheng Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Ludi Li
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - An Zhu
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Wusheng Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing 100191, China; (J.Y.); (S.X.); (L.L.); (A.Z.); (W.X.)
- Key Laboratory of State Administration of Traditional Chinese Medicine (TCM) for Compatibility Toxicology, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
14
|
Singh MK, Shin Y, Ju S, Han S, Kim SS, Kang I. Comprehensive Overview of Alzheimer's Disease: Etiological Insights and Degradation Strategies. Int J Mol Sci 2024; 25:6901. [PMID: 39000011 PMCID: PMC11241648 DOI: 10.3390/ijms25136901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder and affects millions of individuals globally. AD is associated with cognitive decline and memory loss that worsens with aging. A statistical report using U.S. data on AD estimates that approximately 6.9 million individuals suffer from AD, a number projected to surge to 13.8 million by 2060. Thus, there is a critical imperative to pinpoint and address AD and its hallmark tau protein aggregation early to prevent and manage its debilitating effects. Amyloid-β and tau proteins are primarily associated with the formation of plaques and neurofibril tangles in the brain. Current research efforts focus on degrading amyloid-β and tau or inhibiting their synthesis, particularly targeting APP processing and tau hyperphosphorylation, aiming to develop effective clinical interventions. However, navigating this intricate landscape requires ongoing studies and clinical trials to develop treatments that truly make a difference. Genome-wide association studies (GWASs) across various cohorts identified 40 loci and over 300 genes associated with AD. Despite this wealth of genetic data, much remains to be understood about the functions of these genes and their role in the disease process, prompting continued investigation. By delving deeper into these genetic associations, novel targets such as kinases, proteases, cytokines, and degradation pathways, offer new directions for drug discovery and therapeutic intervention in AD. This review delves into the intricate biological pathways disrupted in AD and identifies how genetic variations within these pathways could serve as potential targets for drug discovery and treatment strategies. Through a comprehensive understanding of the molecular underpinnings of AD, researchers aim to pave the way for more effective therapies that can alleviate the burden of this devastating disease.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Cui K, Tang X, Yang B, Fan M, Hu A, Wu P, Yang F, Lin J, Kong H, Lu X, Yu S, Xu Y, Liang X. Sema4D Knockout Attenuates Choroidal Neovascularization by Inhibiting M2 Macrophage Polarization Via Regulation of the RhoA/ROCK Pathway. Invest Ophthalmol Vis Sci 2024; 65:34. [PMID: 38913005 PMCID: PMC11204059 DOI: 10.1167/iovs.65.6.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Purpose The aim of this study was to elucidate the role of Sema4D in the pathogenesis of senescence-associated choroidal neovascularization (CNV) and to explore its underlying mechanisms. Methods In this study, we utilized a model of laser-induced CNV in both young (3 months old) and old (18 months old) mice, including those with or without Sema4D knockout. The expression and localization of Sema4D in CNV were assessed using PCR, Western blot, and immunostaining. Subsequently, the morphological and imaging examinations were used to evaluate the size of CNV and vascular leakage. Finally, the expression of M2 markers, senescence-related markers, and molecules involved in the RhoA/ROCK pathway was detected. Results We found that Sema4D was predominantly expressed in macrophages within CNV lesions, and both the mRNA and protein levels of Sema4D progressively increased following laser photocoagulation, a trend more pronounced in old mice. Moreover, Sema4D knockout markedly inhibited M2 polarization in senescent macrophages and reduced the size and leakage of CNV, particularly in aged mice. Mechanistically, aging was found to upregulate RhoA/ROCK signaling, and knockout of Sema4D effectively suppressed the activation of this pathway, with more significant effects observed in aged mice. Conclusions Our findings revealed that the deletion of Sema4D markedly inhibited M2 macrophage polarization through the suppression of the RhoA/ROCK pathway, ultimately leading to the attenuation of senescence-associated CNV. These data indicate that targeting Sema4D could offer a promising approach for gene editing therapy in patients with neovascular age-related macular degeneration.
Collapse
Affiliation(s)
- Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Matthew Fan
- Yale College, Yale University, New Haven, Connecticut, United States
| | - Andina Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Peiqi Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Fengmei Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Haolin Kong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
16
|
Chiu J, Krupa JM, Seah C, Pasternak SH. Small GTPases control macropinocytosis of amyloid precursor protein and cleavage to amyloid-β. Heliyon 2024; 10:e31077. [PMID: 38799759 PMCID: PMC11126852 DOI: 10.1016/j.heliyon.2024.e31077] [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: 03/21/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The overproduction of the toxic peptide amyloid-beta (Aβ) generated from the cleavage of amyloid precursor protein (APP) is proposed to be a critical event in the development of Alzheimer's disease. Evidence suggests that the cleavage of APP occurs after its internalization from the cell surface. Previously, we identified a novel pathway for APP internalization, which trafficks cell surface APP directly to lysosomes by macropinocytosis, leading to its processing into Aβ. We also demonstrated that ADP-ribosylation factor 6 (Arf6) is required for the macropinocytosis of APP. Here, we characterized the roles of Arf6's downstream effectors Rac1, Cdc42 and RhoA. Both pharmacological inhibition and siRNA knockdown of these proteins reduced the amount of APP colocalized with LAMP1-labeled lysosomes without affecting APP transport to early endosomes. Decreases in the production of both Aβ40 and Aβ42 were also observed by ELISA in response to inhibitor treatment. These findings together demonstrate that Rac1, Cdc42 and RhoA are components of the mechanism regulating the macropinocytosis of APP and targeting these components can reduce the production of Aβ.
Collapse
Affiliation(s)
- Justin Chiu
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jordan M. Krupa
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Claudia Seah
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen H. Pasternak
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
17
|
Santiago-Balmaseda A, Aguirre-Orozco A, Valenzuela-Arzeta IE, Villegas-Rojas MM, Pérez-Segura I, Jiménez-Barrios N, Hurtado-Robles E, Rodríguez-Hernández LD, Rivera-German ER, Guerra-Crespo M, Martinez-Fong D, Ledesma-Alonso C, Diaz-Cintra S, Soto-Rojas LO. Neurodegenerative Diseases: Unraveling the Heterogeneity of Astrocytes. Cells 2024; 13:921. [PMID: 38891053 PMCID: PMC11172252 DOI: 10.3390/cells13110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The astrocyte population, around 50% of human brain cells, plays a crucial role in maintaining the overall health and functionality of the central nervous system (CNS). Astrocytes are vital in orchestrating neuronal development by releasing synaptogenic molecules and eliminating excessive synapses. They also modulate neuronal excitability and contribute to CNS homeostasis, promoting neuronal survival by clearance of neurotransmitters, transporting metabolites, and secreting trophic factors. Astrocytes are highly heterogeneous and respond to CNS injuries and diseases through a process known as reactive astrogliosis, which can contribute to both inflammation and its resolution. Recent evidence has revealed remarkable alterations in astrocyte transcriptomes in response to several diseases, identifying at least two distinct phenotypes called A1 or neurotoxic and A2 or neuroprotective astrocytes. However, due to the vast heterogeneity of these cells, it is limited to classify them into only two phenotypes. This review explores the various physiological and pathophysiological roles, potential markers, and pathways that might be activated in different astrocytic phenotypes. Furthermore, we discuss the astrocyte heterogeneity in the main neurodegenerative diseases and identify potential therapeutic strategies. Understanding the underlying mechanisms in the differentiation and imbalance of the astrocytic population will allow the identification of specific biomarkers and timely therapeutic approaches in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Alberto Santiago-Balmaseda
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Annai Aguirre-Orozco
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Irais E. Valenzuela-Arzeta
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Marcos M. Villegas-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico
| | - Isaac Pérez-Segura
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Natalie Jiménez-Barrios
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Ernesto Hurtado-Robles
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Luis Daniel Rodríguez-Hernández
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Erick R. Rivera-German
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Magdalena Guerra-Crespo
- Laboratorio de Medicina Regenerativa, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico;
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| |
Collapse
|
18
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong DD, Seyfried NT, Wood LB, Rowan MJM, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies unique molecular signatures and vulnerabilities to early Alzheimer's pathology. Nat Commun 2024; 15:2823. [PMID: 38561349 PMCID: PMC10985119 DOI: 10.1038/s41467-024-47028-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Dysfunction in fast-spiking parvalbumin interneurons (PV-INs) may represent an early pathophysiological perturbation in Alzheimer's Disease (AD). Defining early proteomic alterations in PV-INs can provide key biological and translationally-relevant insights. We used cell-type-specific in-vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state PV-IN proteomes. PV-IN proteomic signatures include high metabolic and translational activity, with over-representation of AD-risk and cognitive resilience-related proteins. In bulk proteomes, PV-IN proteins were associated with cognitive decline in humans, and with progressive neuropathology in humans and the 5xFAD mouse model of Aβ pathology. PV-IN CIBOP in early stages of Aβ pathology revealed signatures of increased mitochondria and metabolism, synaptic and cytoskeletal disruption and decreased mTOR signaling, not apparent in whole-brain proteomes. Furthermore, we demonstrated pre-synaptic defects in PV-to-excitatory neurotransmission, validating our proteomic findings. Overall, in this study we present native-state proteomes of PV-INs, revealing molecular insights into their unique roles in cognitive resiliency and AD pathogenesis.
Collapse
Affiliation(s)
- Prateek Kumar
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Annie M Goettemoeller
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Claudia Espinosa-Garcia
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Brendan R Tobin
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Ali Tfaily
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Ruth S Nelson
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Aditya Natu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eric B Dammer
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Juliet V Santiago
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, USA
| | - Sneha Malepati
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lihong Cheng
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Hailian Xiao
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
| | - Duc D Duong
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA
- Department of Biochemistry, Emory University, Atlanta, GA, 30322, USA
| | - Levi B Wood
- Georgia W. Woodruff School of Mechanical Engineering, Parker H. Petit Institute for Bioengineering and Bioscience, and Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30322, USA
- School of Chemical and Biological Engineering, GeoInsrgia titute of Technology, Atlanta, GA, 30322, USA
| | - Matthew J M Rowan
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Srikant Rangaraju
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, USA.
- 3 Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
19
|
Chen XJ, Deng Z, Zhang LL, Pan Y, Fu J, Zou L, Bai Z, Xiao X, Sheng F. Therapeutic potential of the medicinal mushroom Ganoderma lucidum against Alzheimer's disease. Biomed Pharmacother 2024; 172:116222. [PMID: 38310653 DOI: 10.1016/j.biopha.2024.116222] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/06/2024] Open
Abstract
Alzheimer's disease (AD) is a high-incidence neurodegenerative disorder, characterized by cognitive impairment, memory loss, and psychiatric abnormalities. Ganoderma lucidum is a famous medicinal fungus with a long history of dietary intake, containing various bioactive components, and have been documented to exhibit antioxidant, anti-inflammatory, anti-tumor, anti-aging, and immunomodulatory effects, among others. Recent studies have shown that G. lucidum and its components have promising therapeutic potential against AD from various aspects, which can delay the progression of AD, improve cognitive function and quality of life. The underlying mechanisms mainly include inhibiting tau hyperphosphorylation, inhibiting Aβ formation, affecting activated microglia, regulating NF-κB/MAPK signalling pathway, inhibiting neuronal apoptosis, modulating immune system, and inhibiting acetylcholinesterase, etc. This paper systematically reviewed the relevant studies on the therapeutic potential of G. lucidum and its active components for treatment of AD, key points related with the mechanism studies and clinical trials have been discussed, and further perspectives have been proposed. Totally, as a natural medicinal mushroom, G. lucidum has the potential to be developed as effective adjuvant for AD treatment owing to its therapeutic efficacy against multiple pathogenesis of AD. Further mechanical investigation and clinical trials can help unlock the complete potential of G. lucidum as a therapeutic option for AD.
Collapse
Affiliation(s)
- Xu-Jia Chen
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Zhou Deng
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China.
| | - Yan Pan
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Jia Fu
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing 100039, China.
| | - Feiya Sheng
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
20
|
Collu R, Yin Z, Giunti E, Daley S, Chen M, Morin P, Killick R, Wong STC, Xia W. Effect of the ROCK inhibitor fasudil on the brain proteomic profile in the tau transgenic mouse model of Alzheimer's disease. Front Aging Neurosci 2024; 16:1323563. [PMID: 38440100 PMCID: PMC10911083 DOI: 10.3389/fnagi.2024.1323563] [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: 10/18/2023] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction The goal of this study is to explore the pharmacological potential of the amyloid-reducing vasodilator fasudil, a selective Ras homolog (Rho)-associated kinases (ROCK) inhibitor, in the P301S tau transgenic mouse model (Line PS19) of neurodegenerative tauopathy and Alzheimer's disease (AD). Methods We used LC-MS/MS, ELISA and bioinformatic approaches to investigate the effect of treatment with fasudil on the brain proteomic profile in PS19 tau transgenic mice. We also explored the efficacy of fasudil in reducing tau phosphorylation, and the potential beneficial and/or toxic effects of its administration in mice. Results Proteomic profiling of mice brains exposed to fasudil revealed the activation of the mitochondrial tricarboxylic acid (TCA) cycle and blood-brain barrier (BBB) gap junction metabolic pathways. We also observed a significant negative correlation between the brain levels of phosphorylated tau (pTau) at residue 396 and both fasudil and its metabolite hydroxyfasudil. Conclusions Our results provide evidence on the activation of proteins and pathways related to mitochondria and BBB functions by fasudil treatment and support its further development and therapeutic potential for AD.
Collapse
Affiliation(s)
- Roberto Collu
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Zheng Yin
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston Methodist Academic Institute, Houston, TX, United States
| | - Elisa Giunti
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Daley
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Mei Chen
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Peter Morin
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Richard Killick
- King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Stephen T. C. Wong
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston Methodist Academic Institute, Houston, TX, United States
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, United States
| |
Collapse
|
21
|
Haspel N, Jang H, Nussinov R. Allosteric Activation of RhoA Complexed with p115-RhoGEF Deciphered by Conformational Dynamics. J Chem Inf Model 2024; 64:862-873. [PMID: 38215280 DOI: 10.1021/acs.jcim.3c01412] [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: 01/14/2024]
Abstract
The Ras homologue family member A (RhoA) is a member of the Rho family, a subgroup of the Ras superfamily. RhoA interacts with the 115 kDa guanine nucleotide exchange factor (p115-RhoGEF), which assists in activation and binding with downstream effectors. Here, we use molecular dynamics (MD) simulations and essential dynamics analysis of the inactive RhoA-GDP and active RhoA-GTP, when bound to p115-RhoGEF to decipher the mechanism of RhoA activation at the structural level. We observe that inactive RhoA-GDP maintains its position near the catalytic site on the Dbl homology (DH) domain of p115-RhoGEF through the interaction of its Switch I region with the DH domain. We further show that the active RhoA-GTP is engaged in more interactions with the p115-RhoGEF membrane-bound Pleckstrin homology (PH) domain as compared to RhoA-GDP. We hypothesize that the role of the interactions between the active RhoA-GTP and the PH domain is to help release it from the DH domain upon activation. Our results support this premise, and our simulations uncover the beginning of this process and provide structural details. They also point to allosteric communication pathways that take part in RhoA activation to promote and strengthen the interaction between the active RhoA-GTP and the PH domain. Allosteric regulation also occurs among other members of the Rho superfamily. Collectively, we suggest that in the activation process, the role of the RhoA-GTP interaction with the PH domain is to release RhoA-GTP from the DH domain after activation, making it available to downstream effectors.
Collapse
Affiliation(s)
- Nurit Haspel
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts 02125, United States
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
22
|
Ouyang X, Collu R, Benavides GA, Tian R, Darley-Usmar V, Xia W, Zhang J. ROCK Inhibitor Fasudil Attenuates Neuroinflammation and Associated Metabolic Dysregulation in the Tau Transgenic Mouse Model of Alzheimer's Disease. Curr Alzheimer Res 2024; 21:183-200. [PMID: 38910422 DOI: 10.2174/0115672050317608240531130204] [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: 03/18/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND The pathological manifestations of Alzheimer's disease (AD) include not only brain amyloid β protein (Aβ) containing neuritic plaques and hyperphosphorylated tau (p-- tau) containing neurofibrillary tangles but also microgliosis, astrocytosis, and neurodegeneration mediated by metabolic dysregulation and neuroinflammation. METHODS While antibody-based therapies targeting Aβ have shown clinical promise, effective therapies targeting metabolism, neuroinflammation, and p-tau are still an urgent need. Based on the observation that Ras homolog (Rho)-associated kinases (ROCK) activities are elevated in AD, ROCK inhibitors have been explored as therapies in AD models. This study determines the effects of fasudil, a ROCK inhibitor, on neuroinflammation and metabolic regulation in the P301S tau transgenic mouse line PS19 that models neurodegenerative tauopathy and AD. Using daily intraperitoneal (i.p.) delivery of fasudil in PS19 mice, we observed a significant hippocampal-specific decrease of the levels of phosphorylated tau (pTau Ser202/Thr205), a decrease of GFAP+ cells and glycolytic enzyme Pkm1 in broad regions of the brain, and a decrease in mitochondrial complex IV subunit I in the striatum and thalamic regions. RESULTS Although no overt detrimental phenotype was observed, mice dosed with 100 mg/kg/day for 2 weeks exhibited significantly decreased mitochondrial outer membrane and electron transport chain (ETC) protein abundance, as well as ETC activities. CONCLUSION Our results provide insights into dose-dependent neuroinflammatory and metabolic responses to fasudil and support further refinement of ROCK inhibitors for the treatment of AD.
Collapse
Affiliation(s)
- Xiaosen Ouyang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL35294, USA
| | - Roberto Collu
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Gloria A Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL35294, USA
| | - Ran Tian
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL35294, USA
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL35294, USA
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, USA
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL35294, USA
| |
Collapse
|
23
|
Volik PI, Kopeina GS, Zhivotovsky B, Zamaraev AV. Total recall: the role of PIDDosome components in neurodegeneration. Trends Mol Med 2023; 29:996-1013. [PMID: 37716905 DOI: 10.1016/j.molmed.2023.08.008] [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: 07/12/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/18/2023]
Abstract
The PIDDosome is a multiprotein complex that includes p53-induced protein with a death domain 1 (PIDD1), receptor-interacting protein-associated ICH-1/CED-3 homologous protein with a death domain (RAIDD), and caspase-2, the activation of which is driven by PIDDosome assembly. In addition to the key role of the PIDDosome in the regulation of cell differentiation, tissue homeostasis, and organogenesis and regeneration, caspase-2, RAIDD and PIDD1 engagement in neuronal development was shown. Here, we focus on the involvement of PIDDosome components in neurodegenerative disorders, including retinal neuropathies, different types of brain damage, and Alzheimer's disease (AD), Huntington's disease (HD), and Lewy body disease. We also discuss pathogenic variants of PIDD1, RAIDD, and caspase-2 that are associated with intellectual, behavioral, and psychological abnormalities, together with prospective PIDDosome inhibition strategies and their potential clinical application.
Collapse
Affiliation(s)
- Pavel I Volik
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Gelina S Kopeina
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia
| | - Boris Zhivotovsky
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia; Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177 Stockholm, Sweden.
| | - Alexey V Zamaraev
- Facuty of Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia; Engelhardt Institute of Molecular Biology, RAS, 119991 Moscow, Russia.
| |
Collapse
|
24
|
Dai H, Tao S, Guan Y, Zhang Y, Yang Z, Jia J, Zhang X, Zhang G. Astragalus (Astragalus mongholicus) Improves Ventricular Remodeling via ESR1 Downregulation RhoA/ROCK Pathway. Int Heart J 2023; 64:1148-1156. [PMID: 37967985 DOI: 10.1536/ihj.23-265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Astragalus (Astragalus mongholicus) alleviates myocardial remodeling caused by hypertension. However, the detailed molecular mechanism is unclear. This study aims to investigate the effect of Astragalus on ventricular remodeling in ovariectomized spontaneous hypertensive rats (OVX-SHR).Female SHR/NCrl rats were subjected to bilateral ovariectomy to establish the OVX-SHR model and treated with Astragalus extract by gavage. The hemodynamics and cardiac function parameters were measured. HE and Masson staining were used to detect the pathological structure of myocardial remodeling and observe the hyperplasia of myocardial collagen fibers. The immunohistochemistry tested the level of α-SMA. The expression levels of inflammatory cytokines, IκB, p65, Cleaved-Caspase3, RhoA, and ROCK1/2 were detected using Western blot. The method of qRT-PCR measured the expression of matrix metalloproteinase (MMP-2 and MMP-9).Hemodynamic and cardiac function parameters were significantly improved after a high dose of Astragalus extract and Valsartan treatment. The myocardial integrity of the model group was significantly reduced, arranged loosely, and disordered, while the expression of α-SMA was increased. However, Astragalus extract and Valsartan treatments significantly reduced the pathological damage and α-SMA. The levels of TNF-α, IL-1β, IL-6, TGF-β, MMP-2, and MMP-9 in the model group were increased but decreased after Astragalus extract treatment. Adding an ESR1 inhibitor attenuated the improvement effect of Astragalus extract on myocardial remodeling and restored the expression of RhoA and ROCK1/2.Astragalus extract attenuates the cardiac damage in OVX-SHR by downregulating the RhoA/ROCK pathway through ESR1.
Collapse
Affiliation(s)
- Hualei Dai
- Department of Cardiology, The Affiliated Hospital of Yunnan University
- School of Medicine, Yunnan University
| | - Siming Tao
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Yingxia Guan
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Yijian Zhang
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Zhigang Yang
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Ji Jia
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Xinjin Zhang
- Department of Cardiology, The Affiliated Hospital of Yunnan University
| | - Guimin Zhang
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yunnan University
| |
Collapse
|
25
|
Lingli C, Hongmei N, Penghuan J, Hongli Z, Yuye L, Rui W, Fei R, Zhihong Y, Dongfang H, Yaming G. Inhibition of RhoA/ROCK signalling pathway activity improves neural damage and cognitive deficits in the fluorosis model. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115554. [PMID: 37806133 DOI: 10.1016/j.ecoenv.2023.115554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/28/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
Excessive fluoride intake poses health risks to humans and animals. Many studies have indicated that fluoride exposure can damage the cytoskeleton and synapses, which has negative effects on the intellectual development of humans and animals. Our previous study suggested that the RhoA/ROCK signalling pathway is activated by NaF exposure in HT-22 cells and plays a vital role in cytoskeletal assembly and synaptogenesis. However, the mechanism underlying RhoA/ROCK-mediated cytoskeletal injury induced by fluoride remains unclear. In this study, Neuro-2A cells and ICR mice were used to investigate the effects of RhoA/ROCK activation inhibition on NaF-induced synaptic dysfunction and cognitive impairment. We detected the expression of GAP, RhoA, ROCK1/2, and (p)-MLC in vivo and in vitro model. The results showed that NaF exposure activated the RhoA/ROCK/MLC signalling pathway. We measured the effects of RhoA/ROCK inhibition on synaptic injury and intellectual impairment induced by NaF exposure. In vitro, Y-27632 suppressed activated RhoA/ROCK, attenuated morphological and ultrastructural damage, and decreased the survival rate and synapse-functional protein expression caused by NaF. In vivo, the results showed that the RhoA/ROCK/MLC pathway was inhibited by fasudil and improved pathological damage in the hippocampus, cognitive impairment, and decreased expression of neurofunctional proteins induced by NaF. Overall, these results suggest that fasudil and Y-27632 can reverse neurotoxicity caused by fluoride exposure. Furthermore, inhibition of RhoA/ROCK may be a future treatment for CNS injury, and more detailed studies on other neurodegenerative disease models are required to confirm its effectiveness.
Collapse
Affiliation(s)
- Chen Lingli
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China; Postdoctoral Research and Development Base, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ning Hongmei
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Jia Penghuan
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Zhang Hongli
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Liu Yuye
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Wang Rui
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ren Fei
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Yin Zhihong
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Hu Dongfang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China
| | - Ge Yaming
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan Provence, 453003, China.
| |
Collapse
|
26
|
Zhu M, Xiao B, Xue T, Qin S, Ding J, Wu Y, Tang Q, Huang M, Zhao N, Ye Y, Zhang Y, Zhang B, Li J, Guo F, Jiang Y, Zhang L, Zhang L. Cdc42GAP deficiency contributes to the Alzheimer's disease phenotype. Brain 2023; 146:4350-4365. [PMID: 37254741 DOI: 10.1093/brain/awad184] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Alzheimer's disease, the most common cause of dementia, is a chronic degenerative disease with typical pathological features of extracellular senile plaques and intracellular neurofibrillary tangles and a significant decrease in the density of neuronal dendritic spines. Cdc42 is a member of the small G protein family that plays an important role in regulating synaptic plasticity and is regulated by Cdc42GAP, which switches Cdc42 from active GTP-bound to inactive GDP-bound states regulating downstream pathways via effector proteins. However, few studies have focused on Cdc42 in the progression of Alzheimer's disease. In a heterozygous Cdc42GAP mouse model that exhibited elevated Cdc42-GTPase activity accompanied by increased Cdc42-PAK1-cofilin signalling, we found impairments in cognitive behaviours, neuron senescence, synaptic loss with depolymerization of F-actin and the pathological phenotypes of Alzheimer's disease, including phosphorylated tau (p-T231, AT8), along with increased soluble and insoluble Aβ1-42 and Aβ1-40, which are consistent with typical Alzheimer's disease mice. Interestingly, these impairments increased significantly with age. Furthermore, the results of quantitative phosphoproteomic analysis of the hippocampus of 11-month-old GAP mice suggested that Cdc42GAP deficiency induces and accelerates Alzheimer's disease-like phenotypes through activation of GSK-3β by dephosphorylation at Ser9, Ser389 and/or phosphorylation at Tyr216. In addition, overexpression of dominant-negative Cdc42 in the primary hippocampal and cortical neurons of heterozygous Cdc42GAP mice reversed synaptic loss and tau hyperphosphorylation. Importantly, the Cdc42 signalling pathway, Aβ1-42, Aβ1-40 and GSK-3β activity were increased in the cortical sections of Alzheimer's disease patients compared with those in healthy controls. Together, these data indicated that Cdc42GAP is involved in regulating Alzheimer's disease-like phenotypes such as cognitive deficits, dendritic spine loss, phosphorylated tau (p-T231, AT8) and increased soluble and insoluble Aβ1-42 and Aβ1-40, possibly through the activation of GSK-3β, and these impairments increased significantly with age. Thus, we provide the first evidence that Cdc42 is involved in the progression of Alzheimer's disease-like phenotypes, which may provide new targets for Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Mengjuan Zhu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Xiao
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Tao Xue
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Sifei Qin
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jiuyang Ding
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yue Wu
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Qingqiu Tang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Mengfan Huang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Na Zhao
- School of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yingshan Ye
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuning Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Boya Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Juan Li
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Center for Orthopedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Research Foundation, Cincinnati, OH 45229-3026, USA
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin Zhang
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Center for Orthopedic Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Functional Proteomics, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Department of Otorhinolaryngology-Head and Neck Surgery of the Third Affiliated Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
27
|
Benarroch E. What Is the Role of the Rho-ROCK Pathway in Neurologic Disorders? Neurology 2023; 101:536-543. [PMID: 37722862 PMCID: PMC10516277 DOI: 10.1212/wnl.0000000000207779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 09/20/2023] Open
|
28
|
Wu Y, Yang Y, Liu J, Li Y, Pi R, Ren Y, Jiang T, Wang Y, Zhong G. Pharmacokinetic and safety profile of PT109B, a novel multi-targeted compound against Alzheimer's disease. Eur J Pharm Sci 2023; 188:106532. [PMID: 37479046 DOI: 10.1016/j.ejps.2023.106532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/23/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
PT109B, 5-(1,2-dithiolan-3-yl)-N-((1r,4r)-4-(isoquinolin-5-ylamino) cyclohexyl) pentanamide, a novel compound structurally related to Fasudil, has been reported as a promising candidate for treating Alzheimer's disease. To investigate the pharmacokinetics and acute toxicity of PT109B in rodents, we first developed and validated a UPLC-MS/MS analytical method to detect PT109B concentration in the biological matrix. The proposed method could separate and quantify the PT109B with good precision and accuracy. The pharmacokinetic results showed that the concentrations of PT109B in rat plasma increased with the dose, but not proportionally. Meanwhile, the double-peak phenomenon disappeared when decreasing the oral administration dosage. In addition, we found that PT109B could be detected in the central nervous system, and highly distributed in the liver and kidney. At the same time, the gender difference of PT109B in rats was observed, and the exposure of PT109B in female rats was significantly higher than that in male rats after oral administration. Finally, we found that oral administration of 750 mg/kg PT109B to C57 BL/6 mice caused significant liver injury in females, which was specifically manifested as hepatomegaly, increased liver coefficient, and hepatocyte ballooning. However, no significant damage was observed in other organs, which may be related to the distribution of PT109B in the liver. In summary, we first established a UPLC-MS/MS method for the analysis of PT109B in a biological matrix and described the characteristics of pharmacokinetics, and acute toxicity of PT109B in rodents, providing a sufficient pharmacokinetic basis for further study of PT109B.
Collapse
Affiliation(s)
- Yufeng Wu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Yang Yang
- School of Medicine, Shenzhen campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Jingyu Liu
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Yagang Li
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Rongbiao Pi
- School of Medicine, Shenzhen campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yu Ren
- School of Medicine, Shenzhen campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Tianyang Jiang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Yuran Wang
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Guoping Zhong
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510000, China; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
29
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
30
|
Huang YR, Xie XX, Yang J, Sun XY, Niu XY, Yang CG, Li LJ, Zhang L, Wang D, Liu CY, Hou SJ, Jiang CY, Xu YM, Liu RT. ArhGAP11A mediates amyloid-β generation and neuropathology in an Alzheimer's disease-like mouse model. Cell Rep 2023; 42:112624. [PMID: 37302068 DOI: 10.1016/j.celrep.2023.112624] [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: 06/13/2022] [Revised: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Amyloid-β (Aβ) plays an important role in the neuropathology of Alzheimer's disease (AD), but some factors promoting Aβ generation and Aβ oligomer (Aβo) neurotoxicity remain unclear. We here find that the levels of ArhGAP11A, a Ras homology GTPase-activating protein, significantly increase in patients with AD and amyloid precursor protein (APP)/presenilin-1 (PS1) mice. Reducing the ArhGAP11A level in neurons not only inhibits Aβ generation by decreasing the expression of APP, PS1, and β-secretase (BACE1) through the RhoA/ROCK/Erk signaling pathway but also reduces Aβo neurotoxicity by decreasing the expressions of apoptosis-related p53 target genes. In APP/PS1 mice, specific reduction of the ArhGAP11A level in neurons significantly reduces Aβ production and plaque deposition and ameliorates neuronal damage, neuroinflammation, and cognitive deficits. Moreover, Aβos enhance ArhGAP11A expression in neurons by activating E2F1, which thus forms a deleterious cycle. Our results demonstrate that ArhGAP11A may be involved in AD pathogenesis and that decreasing ArhGAP11A expression may be a promising therapeutic strategy for AD treatment.
Collapse
Affiliation(s)
- Ya-Ru Huang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xi-Xiu Xie
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Yang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Henan Medical Key Laboratory of Neurogenetic and Neurodegenerative Disease, Zhengzhou 450052, Henan, China
| | - Xiao-Ying Sun
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Yun Niu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Ningxia University, Yinchuan 750021, Ningxia, China
| | - Cheng-Gang Yang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China; Department of Research and Development, Gu'an Bojian Bio-Technology Co., Ltd., Langfang 065000, Hebei, China
| | - Ling-Jie Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lun Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Dan Wang
- Department of BigData, Beijing Medintell Bioinformatic Technology Co., Ltd., Beijing 100081, China
| | - Chun-Yu Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; Shandong Agricultural University, Tai'an 271000, Shandong, China
| | - Sheng-Jie Hou
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen-Yang Jiang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Key Laboratory of Cerebrovascular Disease of Henan Province, Zhengzhou 450052, Henan, China.
| | - Rui-Tian Liu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
31
|
Kumar P, Goettemoeller AM, Espinosa-Garcia C, Tobin BR, Tfaily A, Nelson RS, Natu A, Dammer EB, Santiago JV, Malepati S, Cheng L, Xiao H, Duong D, Seyfried NT, Wood LB, Rowan MJ, Rangaraju S. Native-state proteomics of Parvalbumin interneurons identifies novel molecular signatures and metabolic vulnerabilities to early Alzheimer's disease pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541038. [PMID: 37292756 PMCID: PMC10245729 DOI: 10.1101/2023.05.17.541038] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the earliest pathophysiological perturbations in Alzheimer's Disease (AD) may arise from dysfunction of fast-spiking parvalbumin (PV) interneurons (PV-INs). Defining early protein-level (proteomic) alterations in PV-INs can provide key biological and translationally relevant insights. Here, we use cell-type-specific in vivo biotinylation of proteins (CIBOP) coupled with mass spectrometry to obtain native-state proteomes of PV interneurons. PV-INs exhibited proteomic signatures of high metabolic, mitochondrial, and translational activity, with over-representation of causally linked AD genetic risk factors. Analyses of bulk brain proteomes indicated strong correlations between PV-IN proteins with cognitive decline in humans, and with progressive neuropathology in humans and mouse models of Aβ pathology. Furthermore, PV-IN-specific proteomes revealed unique signatures of increased mitochondrial and metabolic proteins, but decreased synaptic and mTOR signaling proteins in response to early Aβ pathology. PV-specific changes were not apparent in whole-brain proteomes. These findings showcase the first native state PV-IN proteomes in mammalian brain, revealing a molecular basis for their unique vulnerabilities in AD.
Collapse
|
32
|
Mankhong S, Kim S, Moon S, Choi SH, Kwak HB, Park DH, Shah P, Lee PH, Yang SW, Kang JH. Circulating micro-RNAs Differentially Expressed in Korean Alzheimer's Patients With Brain Aβ Accumulation Activate Amyloidogenesis. J Gerontol A Biol Sci Med Sci 2023; 78:292-303. [PMID: 35532940 DOI: 10.1093/gerona/glac106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Roles for extracellular vesicles (EVs) enriched with micro-RNAs (miRNAs) have been proposed in Alzheimer's disease (AD) pathogenesis, leading to the discovery of blood miRNAs as AD biomarkers. However, the diagnostic utility of specific miRNAs is not consistent. This study aimed to discover blood miRNAs that are differentially expressed in Korean AD patients, evaluate their clinical performance, and investigate their role in amyloidogenesis. METHODS We discovered miRNAs differentially expressed in AD (N = 8) from cognitively normal participants (CN, N = 7) or Parkinson's disease (PD) patients (N = 8). We evaluated the clinical performance of these miRNAs in plasma of subgroup (N = 99) and in plasma EVs isolated from the total cohort (N = 251). The effects of miRNAs on amyloidogenesis and on the regulation of their target genes were investigated in vitro. RESULTS Among 17 upregulated and one downregulated miRNAs in AD (>twofold), miR-122-5p, miR-210-3p, and miR-590-5p were differentially expressed compared with CN or PD. However, the diagnostic performance of the selected plasma or EV miRNAs in total participants were limited (area under the curve < 0.8). Nevertheless, levels of 3 miRNAs in plasma or plasma EVs of participants who were amyloid positron emission tomography (Aβ-PET) positive were significantly higher than those from the Aβ-PET negative participants (p < .05). The selected miRNAs induced Aβ production (p < .05) through activation of β-cleavage of amyloid precursor protein (CTF-β; p < .01), and downregulated their target genes (ADAM metallopeptidase domain 10, Brain-derived neurotrophic factor, and Jagged canonical notch ligand 1; p < .05), which was further supported by pathway enrichment analysis of target genes of the miRNAs. CONCLUSION In conclusion, despite of the limited diagnostic utility of selected miRNAs as plasma or plasma EV biomarkers, the discovered miRNAs may play a role in amyloidogenesis during AD onset and progression.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, Republic of Korea.,Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| | - Sujin Kim
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Sohee Moon
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Seong-Hye Choi
- Department of Neurology, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hyo-Bum Kwak
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea.,Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Dong-Ho Park
- Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea.,Department of Kinesiology, Inha University, Incheon, Republic of Korea
| | - Pratik Shah
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seong Wook Yang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Research Center for Controlling Intercellular Communication, College of Medicine, Inha University, Incheon, Republic of Korea.,Program in Biomedical Science and Engineering, Inha University, Incheon, South Korea
| |
Collapse
|
33
|
Feng G, Liu X, Wang B, Li R, Chang Y, Guo N, Li Y, Chen T, Ma B. Exploring the mechanism of Chaihujia Longgu Muli decoction in the treatment of epilepsy in rats based on the RhoA/ROCK signaling pathway. Mol Biol Rep 2023; 50:3389-3399. [PMID: 36739316 DOI: 10.1007/s11033-023-08301-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/23/2023] [Indexed: 02/06/2023]
Abstract
BACKGROUND The Chinese herbal formula Chaihujia Longgu Muli Decoction (CD) has a good antiepileptic effect, but its mechanisms remain unclear. Therefore, in this study we explored the molecular mechanisms of CD against epilepsy. METHODS Twelve-day-old SD rats were randomly divided into a normal group, model group, valproic acid group, and CD high, medium, and low groups. Except for the normal group, the other groups were given an intraperitoneal injection of pentylenetetrazol (PTZ) to establish epilepsy models, and the Racine score was applied for model judgment. After 14 consecutive days of dosing, the Morris water maze test was performed. Then, hippocampal Nissl staining and immunofluorescence staining were performed, and synaptic ultrastructure was observed by transmission electron microscopy (TEM). RhoA/ROCK signaling pathway proteins were detected. RESULTS In PTZ model rats, the passing times were reduced, and the escape latency was prolonged in the Morris water maze test. Nissl staining showed that some hippocampal neurons swelled and ruptured, Nissl bodies in the cytoplasm were significantly reduced, and neurons were lost. Immunofluorescence detection revealed that the expression of PSD95 and SYP was significantly reduced. Electron microscopy results revealed that the number of synapses in hippocampal neurons was significantly reduced and the postsynaptic membrane length was significantly reduced. Western blot analysis showed that the RhoA/ROCK signaling pathway was activated, while SYP, SPD95, and PTEN expression was significantly decreased. After treatment with CD, neurobehavioral abnormalities and neuronal damage caused by epileptic seizures were improved. CONCLUSION CD exerted an antiepileptic effect by inhibiting the activation of the RhoA/ROCK signaling pathway.
Collapse
Affiliation(s)
- Gang Feng
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xianghua Liu
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Baoying Wang
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ruixing Li
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yaxin Chang
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Nannan Guo
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yawei Li
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Tiantian Chen
- The First Affiliated Hospital of Henan University of Chinese Medicine, Shanghai, 450099, China
| | - Bingxiang Ma
- College of Pediatric Medicine, Henan University of Chinese Medicine, Zhengzhou, 450046, China. .,The First Affiliated Hospital of Henan University of Chinese Medicine, Shanghai, 450099, China. .,, No. 19, Renmin Road, Jinshui District, Zhengzhou, 450099, China.
| |
Collapse
|
34
|
Investigating the Anticancer Activity of G-Rh1 Using In Silico and In Vitro Studies (A549 Lung Cancer Cells). MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238311. [PMID: 36500403 PMCID: PMC9890317 DOI: 10.3390/molecules27238311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022]
Abstract
Ginsenoside Rh1 (G-Rh1), a possible bioactive substance isolated from the Korean Panax ginseng Meyer, has a wide range of pharmacological effects. In this study, we have investigated the anticancer efficacy of G-Rh1 via in silico and in vitro methodologies. This study mainly focuses on the two metastatic regulators, Rho-associated protein kinase 1 (ROCK1) and RhoA, along with other standard apoptosis regulators. The ROCK1 protein is a member of the active serine/threonine kinase family that is crucial for many biological processes, including cell division, differentiation, and death, as well as many cellular processes and muscle contraction. The abnormal activation of ROCK1 kinase causes several disorders, whereas numerous studies have also shown that RhoA is expressed highly in various cancers, including colon, lung, ovarian, gastric, and liver malignancies. Hence, inhibiting both ROCK1 and RhoA will be promising in preventing metastasis. Therefore, the molecular level interaction of G-Rh1 with the ROCK1 and RhoA active site residues from the preliminary screening clearly shows its inhibitory potential. Molecular dynamics simulation and principal component analysis give essential insights for comprehending the conformational changes that result from G-Rh1 binding to ROCK1 and RhoA. Further, MTT assay was employed to examine the potential cytotoxicity in vitro against human lung cancer cells (A549) and Raw 264.7 Murine macrophage cells. Thus, G-Rh1 showed significant cytotoxicity against human lung adenocarcinoma (A549) at 100 µg/mL. In addition, we observed an elevated level of reactive oxygen species (ROS) generation, perhaps promoting cancer cell toxicity. Additionally, G-Rh1 suppressed the mRNA expression of RhoA, ROCK1, MMP1, and MMP9 in cancer cell. Accordingly, G-Rh1 upregulated the p53, Bax, Caspase 3, caspase 9 while Bcl2 is downregulated intrinsic pathway. The findings from our study propose that the anticancer activity of G-Rh1 may be related to the induction of apoptosis by the RhoA/ROCK1 signaling pathway. As a result, this study evaluated the functional drug-like compound G-Rh1 from Panax ginseng in preventing and treating lung cancer adenocarcinoma via regulating metastasis and apoptosis.
Collapse
|
35
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
36
|
Multitargeting the Action of 5-HT 6 Serotonin Receptor Ligands by Additional Modulation of Kinases in the Search for a New Therapy for Alzheimer's Disease: Can It Work from a Molecular Point of View? Int J Mol Sci 2022; 23:ijms23158768. [PMID: 35955902 PMCID: PMC9368844 DOI: 10.3390/ijms23158768] [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: 07/20/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022] Open
Abstract
In view of the unsatisfactory treatment of cognitive disorders, in particular Alzheimer’s disease (AD), the aim of this review was to perform a computer-aided analysis of the state of the art that will help in the search for innovative polypharmacology-based therapeutic approaches to fight against AD. Apart from 20-year unrenewed cholinesterase- or NMDA-based AD therapy, the hope of effectively treating Alzheimer’s disease has been placed on serotonin 5-HT6 receptor (5-HT6R), due to its proven, both for agonists and antagonists, beneficial procognitive effects in animal models; however, research into this treatment has so far not been successfully translated to human patients. Recent lines of evidence strongly emphasize the role of kinases, in particular microtubule affinity-regulating kinase 4 (MARK4), Rho-associated coiled-coil-containing protein kinase I/II (ROCKI/II) and cyclin-dependent kinase 5 (CDK5) in the etiology of AD, pointing to the therapeutic potential of their inhibitors not only against the symptoms, but also the causes of this disease. Thus, finding a drug that acts simultaneously on both 5-HT6R and one of those kinases will provide a potential breakthrough in AD treatment. The pharmacophore- and docking-based comprehensive literature analysis performed herein serves to answer the question of whether the design of these kind of dual agents is possible, and the conclusions turned out to be highly promising.
Collapse
|
37
|
Effects of RhoA on depression-like behavior in prenatally stressed offspring rats. Behav Brain Res 2022; 432:113973. [PMID: 35728732 DOI: 10.1016/j.bbr.2022.113973] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022]
Abstract
Depression is a common mental disease that can lead to suicide when severe. Exposure to prenatal stress (PS) can lead to depression-like behavior in offspring, but the mechanism is unclear. RhoA (Ras homology family member A) plays an important role in stress-induced changes in synaptic plasticity, participating in the development of depression by activating the downstream effector ROCK (Rho-associated protein kinase). This study explored the influence in the expression of RhoA and downstream molecules ROCK1/2 in prenatally stressed rats, and the effect of RhoA inhibitor simvastatin on depression-like behavior induced by PS. Depression-like behavior in offspring was detected by sucrose preference test, forced swimming test, and open-field test. The mRNA and protein expression of RhoA and ROCK1/2 in the hippocampus and prefrontal cortex of offspring rats were detected by qRT-PCR and western blotting, respectively. Our results showed that PS causes depression-like behavior in offspring rats, associated with elevated expression of RhoA, ROCK1/2 in the hippocampus and prefrontal cortex. After administration of simvastatin to PS rats, the expression of RhoA and ROCK2 was significantly reduced, alleviating depression-like behavior. Our study demonstrated that RhoA participates in the depression-like behavior in prenatally stressed offspring rats, which may be a potential target for antidepressant therapy.
Collapse
|
38
|
RhoA Signaling in Neurodegenerative Diseases. Cells 2022; 11:cells11091520. [PMID: 35563826 PMCID: PMC9103838 DOI: 10.3390/cells11091520] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023] Open
Abstract
Ras homolog gene family member A (RhoA) is a small GTPase of the Rho family involved in regulating multiple signal transduction pathways that influence a diverse range of cellular functions. RhoA and many of its downstream effector proteins are highly expressed in the nervous system, implying an important role for RhoA signaling in neurons and glial cells. Indeed, emerging evidence points toward a role of aberrant RhoA signaling in neurodegenerative diseases such as Parkinson’s disease, Alzheimer’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. In this review, we summarize the current knowledge of RhoA regulation and downstream cellular functions with an emphasis on the role of RhoA signaling in neurodegenerative diseases and the therapeutic potential of RhoA inhibition in neurodegeneration.
Collapse
|
39
|
Zhang C, Lin Y, Yan CH, Zhang W. Adipokine Signaling Pathways in Osteoarthritis. Front Bioeng Biotechnol 2022; 10:865370. [PMID: 35519618 PMCID: PMC9062110 DOI: 10.3389/fbioe.2022.865370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease that affects millions of individuals. The pathogenesis of OA has not been fully elucidated. Obesity is a well-recognized risk factor for OA. Multiple studies have demonstrated adipokines play a key role in obesity-induced OA. Increasing evidence show that various adipokines may significantly affect the development or clinical course of OA by regulating the pro/anti-inflammatory and anabolic/catabolic balance, matrix remodeling, chondrocyte apoptosis and autophagy, and subchondral bone sclerosis. Several signaling pathways are involved but still have not been systematically investigated. In this article, we review the cellular and molecular mechanisms of adipokines in OA, and highlight the possible signaling pathways. The review suggested adipokines play important roles in obesity-induced OA, and exert downstream function via the activation of various signaling pathways. In addition, some pharmaceuticals targeting these pathways have been applied into ongoing clinical trials and showed encouraging results. However, these signaling pathways are complex and converge into a common network with each other. In the future work, more research is warranted to further investigate how this network works. Moreover, more high quality randomised controlled trials are needed in order to investigate the therapeutic effects of pharmaceuticals against these pathways for the treatment of OA. This review may help researchers to better understand the pathogenesis of OA, so as to provide new insight for future clinical practices and translational research.
Collapse
Affiliation(s)
- Chaofan Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yunzhi Lin
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chun Hoi Yan
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wenming Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
40
|
Lapresa R, Agulla J, Gonzalez-Guerrero S, Bolaños JP, Almeida A. Amyloid-β Induces Cdh1-Mediated Rock2 Stabilization Causing Neurodegeneration. Front Pharmacol 2022; 13:884470. [PMID: 35496276 PMCID: PMC9047900 DOI: 10.3389/fphar.2022.884470] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/01/2022] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, which is causally related to the accumulation of abnormally folded amyloid-β (Aβ) peptide and hyperphosphorylated tau protein aggregates. The dendritic spine regulator Rho protein kinase 2 (Rock2) accumulates in the brain at the earliest stages of AD and remains increased during disease progression. However, the molecular mechanism that upregulates Rock2 in AD, and its role in the disease progression, are unknown. Here, we found that oligomers of the amyloidogenic fragment 25–35 of the Aβ peptide (Aβ25-35) trigger Rock2 accumulation and activation in mouse cortical neurons in primary culture and in mouse hippocampus in vivo. Neuronal apoptotic death and memory impairment caused by Aβ25-35 administration were rescued by genetic and pharmacological inhibition of Rock2 activity. Mechanistically, Aβ25-35 elicited cyclin dependent kinase-5 (Cdk5)-mediated phosphorylation of Cdh1, a cofactor that is essential for the activity of the E3 ubiquitin ligase anaphase-promoting complex/cyclosome (APC/C) in neurons. Notably, phosphorylated Cdh1 was disassembled from the APC/C complex, causing its inactivation and subsequent Rock2 protein stabilization and activation. Moreover, Aβ25-35-induced neuronal apoptosis was prevented by expressing a phosphodefective form of Cdh1, but not by a phosphomimetic Cdh1. Finally, Cdh1 inactivation, using both genetic and pharmacological approaches, enhanced Aβ25-35-mediated neuronal death through a mechanism that was prevented by inhibition of Rock2 activity. These results indicate that the Cdk5-Cdh1 signaling pathway accounts for the increased Rock2 activity by amyloidogenic Aβ peptides and that this mechanism may contribute to neurodegeneration and memory loss in AD.
Collapse
Affiliation(s)
- Rebeca Lapresa
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, CSIC, University of Salamanca, Salamanca, Spain
| | - Jesus Agulla
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, CSIC, University of Salamanca, Salamanca, Spain
| | - Sonia Gonzalez-Guerrero
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, CSIC, University of Salamanca, Salamanca, Spain
| | - Juan P. Bolaños
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, CSIC, University of Salamanca, Salamanca, Spain
| | - Angeles Almeida
- Institute of Functional Biology and Genomics, CSIC, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, CSIC, University of Salamanca, Salamanca, Spain
- *Correspondence: Angeles Almeida,
| |
Collapse
|
41
|
Zhu Z, Lu J, Wang S, Peng W, Yang Y, Chen C, Zhou X, Yang X, Xin W, Chen X, Pi J, Yin W, Yao L, Pi R. Acrolein, an endogenous aldehyde induces synaptic dysfunction in vitro and in vivo: Involvement of RhoA/ROCK2 pathway. Aging Cell 2022; 21:e13587. [PMID: 35315217 PMCID: PMC9009232 DOI: 10.1111/acel.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/19/2022] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Acrolein, an unsaturated aldehyde, is increased in the brain of Alzheimer's disease (AD) patients and identified as a potential inducer of sporadic AD. Synaptic dysfunction, as a typical pathological change occurring in the early stage of AD, is most closely associated with the severity of dementia. However, there remains a lack of clarity on the mechanisms of acrolein inducing AD-like pathology and synaptic impairment. In this study, acrolein-treated primary cultured neurons and mice were applied to investigate the effects of acrolein on cognitive impairment and synaptic dysfunction and their signaling mechanisms. In vitro, ROCK inhibitors, Fasudil, and Y27632, could attenuate the axon ruptures and synaptic impairment caused by acrolein. Meanwhile, RNA-seq distinct differentially expressed genes in acrolein models and initially linked activated RhoA/Rho-kinase2 (ROCK2) to acrolein-induced synaptic dysfunction, which could regulate neuronal cytoskeleton and neurite. The Morris water maze test and in vivo field excitatory postsynaptic potential (fEPSP) were performed to evaluate spatial memory and long-term potential (LTP), respectively. Acrolein induced cognitive impairment and attenuated LTP. Furthermore, the protein level of Synapsin 1 and postsynaptic density 95 (PSD95) and dendritic spines density were also decreased in acrolein-exposed mice. These changes were improved by ROCK2 inhibitor Fasudil or in ROCK2+/- mice. Together, our findings suggest that RhoA/ROCK2 signaling pathway plays a critical role in acrolein-induced synaptic damage and cognitive dysfunction, suggesting inhibition of ROCK2 should benefit to the early AD.
Collapse
Affiliation(s)
- Zeyu Zhu
- School of Medicine Sun Yat‐Sen University Guangzhou China
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Junfeng Lu
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
- Department of Internal Medicine The Affiliated Tumor Hospital of Zhengzhou University Zhengzhou China
| | - Shuyi Wang
- School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Weijia Peng
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Yang Yang
- School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Chen Chen
- School of Pharmaceutical Sciences Sun Yat‐Sen University Guangzhou China
| | - Xin Zhou
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen Center for Disease Control and Prevention
| | - Wenjun Xin
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Xinyi Chen
- School of Pharmaceutical Sciences South China Research Center for Acupuncture and Moxibustion Guangzhou University of Chinese Medicine Guangzhou China
| | - Jiakai Pi
- Guangzhou Foreign Language School Guangzhou China
| | - Wei Yin
- Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Lin Yao
- Research Institute of Acupuncture and Moxibustion Shandong University of Traditional Chinese Medicine Jinan China
| | - Rongbiao Pi
- School of Medicine Sun Yat‐Sen University Guangzhou China
- International Joint Laboratory<SYSU‐PolyU HK> of Novel Anti‐Dementia Drugs of Guangzhou Guangzhou China
- Guangdong Province Key Laboratory of Brain Function and Disease Sun Yat‐sen University Guangzhou China
| |
Collapse
|
42
|
Xiang X, Wang X, Jin S, Hu J, Wu Y, Li Y, Wu X. Activation of GPR55 attenuates cognitive impairment and neurotoxicity in a mouse model of Alzheimer's disease induced by Aβ 1-42 through inhibiting RhoA/ROCK2 pathway. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110423. [PMID: 34363866 DOI: 10.1016/j.pnpbp.2021.110423] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/11/2021] [Accepted: 07/31/2021] [Indexed: 12/17/2022]
Abstract
The accumulation of amyloid-β (Aβ) peptides in the brain is considered to be the initial event in the Alzheimer's disease (AD). Neurotoxicity mediated by Aβ has been demonstrated to damage the cognitive function. In the present study, we sought to determine the effects of O-1602, a specific G-protein coupled receptor 55 (GPR55) agonist, on the impairment of learning and memory induced by intracerebroventricular (i.c.v.) of Aβ1-42 (400 pmol/mouse) in mice. Our results showed that i.c.v. injection of aggregated Aβ1-42 into the brain of mice resulted in cognitive impairment and neurotoxicity. In contrast, O-1602 (2.0 or 4.0 μg/mouse, i.c.v.) can improve memory impairment induced by Aβ1-42 in the Morris water maze (MWM), and novel object recognition (NOR) tests. Besides, we found that O-1602 reduced the activity of β-secretase 1 (BACE1) and the level of soluble Aβ1-42 in the hippocampus and frontal cortex. Importantly, O-1602 treatment reversed Aβ1-42-induced GPR55 down-regulation, decreased pro-inflammatory cytokines, and the level of malondialdehyde (MDA), increased the levels of glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT), as well as suppressed apoptosis as indicated by decreased TUNEL-positive cells, and increased the ratio of Bcl-2/Bax. O-1602 treatment also pronouncedly ameliorated synaptic dysfunction by promoting the upregulation of PSD-95 and synaptophysin (SYN) proteins. Moreover, O-1602 concurrently down regulated the protein levels of RhoA, and ROCK2, the critical proteins in the RhoA/ROCK2 pathway. This study indicates that O-1602 may reverse Aβ1-42-induced cognitive impairment and neurotoxicity in mice by inhibiting RhoA/ROCK2 pathway. Taken together, these findings suggest that GPR55 could be a novel and promising target for the treatment of AD.
Collapse
Affiliation(s)
- XiaoTong Xiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Xin Wang
- West Anhui Health Vocational College, Luan 237000, China
| | - ShiYu Jin
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Jie Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - YuMei Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - YueYue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China
| | - Xian Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China.
| |
Collapse
|
43
|
Activation of GPR55 attenuates cognitive impairment, oxidative stress, neuroinflammation, and synaptic dysfunction in a streptozotocin-induced Alzheimer's mouse model. Pharmacol Biochem Behav 2022; 214:173340. [DOI: 10.1016/j.pbb.2022.173340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 12/20/2021] [Accepted: 01/20/2022] [Indexed: 12/14/2022]
|
44
|
Chen C, Lu J, Peng W, Mak MS, Yang Y, Zhu Z, Wang S, Hou J, Zhou X, Xin W, Hu Y, Tsim KWK, Han Y, Liu Q, Pi R. Acrolein, an endogenous aldehyde induces Alzheimer's disease-like pathologies in mice: A new sporadic AD animal model. Pharmacol Res 2022; 175:106003. [PMID: 34838693 DOI: 10.1016/j.phrs.2021.106003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/01/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that mainly affects elderly people. However, the translational research of AD is frustrating, suggesting that the development of new AD animal models is crucial. By gavage administration of acrolein, we constructed a simple sporadic AD animal model which showed classic pathologies of AD in 1 month. The AD-like phenotypes and pathological changes were as followed. 1) olfactory dysfunctions, cognitive impairments and psychological symptoms in C57BL/6 mice; 2) increased levels of Aβ1-42 and Tau phosphorylation (S396/T231) in cortex and hippocampus; 3) astrocytes and microglia proliferation; 4) reduced levels of postsynaptic density 95(PSD95) and Synapsin1, as well as the density of dendritic spines in the CA1 and DG neurons of the hippocampus; 5) high-frequency stimulation failed to induce the long-term potentiation (LTP) in the hippocampus after exposure to acrolein for 4 weeks; 6) decreased blood oxygen level-dependent (BOLD) signal in the olfactory bulb and induced high T2 signals in the hippocampus, which matched to the clinical observation in the brain of AD patients, and 7) activated RhoA/ROCK2/ p-cofilin-associated pathway in hippocampus of acrolein-treated mice, which may be the causes of synaptic damage and neuroinflammation in acrolein mice model. Taken together, the acrolein-induced sporadic AD mouse model closely reflects the pathological features of AD, which will be useful for the research on the mechanism of AD onset and the development of anti-AD drugs.
Collapse
Affiliation(s)
- Chen Chen
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Junfeng Lu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Weijia Peng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Marvin Sh Mak
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yang Yang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Zeyu Zhu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shuyi Wang
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Jiawei Hou
- Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xin Zhou
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wenjun Xin
- Zhongshan school of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou 510014, China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese Medicine, The Hong Kong Polytechnic University, Hong Kong, China
| | - Qinyu Liu
- The seventh affiliated hospital, Sun Yat-Sen University, Shenzhen 518107, China.
| | - Rongbiao Pi
- Department of Pharmacology, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangzhou, Guangzhou 510006, China; Neurobiology Research Center, School of Medicine, Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|