1
|
Zhang W, Teng F, Lan X, Liu P, Wang A, Zhang F, Cui Z, Guan J, Sun X. A novel finding relates to the involvement of ATF3/DOCK8 in Alzheimer's disease pathogenesis. J Alzheimers Dis 2025:13872877251336266. [PMID: 40267290 DOI: 10.1177/13872877251336266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe involvement of microglia is likely to be pivotal in the pathogenesis of Alzheimer's disease (AD) by modulating the deposition of amyloid-β (Aβ) plaques. The deletion of Dedicator of cytokinesis 8 (DOCK8) has a protective effect in mouse with neurodegenerative diseases.ObjectiveTo explore the underlying mechanism of DOCK8 in AD.MethodsIn present study, we first the detected the expression of DOCK8 in the hippocampal tissue of APP/PS1 mice. Then, the expression of DOCK8 was knocked down in the hippocampal tissue of APP/PS1 mice, and the effects of DOCK8 down-regulation on cognitive function, the microglia migration around Aβ plaques, and the cell division cycle 42 (Cdc42)/p38 mitogen-activated protein kinase (MAPK) signaling pathway were detected. Next, the effects of DOCK8 knockdown on Aβ-induced migration and activation of BV-2 cells as well as the MAPK signaling pathway were detected. Finally, the transcriptional regulation of DOCK by transcription factor 3 (ATF3) was detected by a dual luciferase reporter assay.ResultsDOCK8 expression exerts a significant upregulation in the hippocampus of APP/PS1 mice. However, following the DOCK8 knockdown, there was a significant recovery in the results of the behavioral tests and a notable reduction in microglial expression. Moreover, the high expression of DOCK8 mediated by ATF3 successfully triggered the Cdc42/p38 MAPK signaling pathway, thereby enhancing the migration and recruitment of microglia towards senile plaques, accelerating the production of Aβ plaques.ConclusionsATF3-mediated high expression of DOCK8 accelerates the production of Aβ plaques, and participates in the pathogenesis of AD.
Collapse
Affiliation(s)
- Wenqiang Zhang
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fei Teng
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xifa Lan
- Department of Neurology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, People's Republic of China
| | - Peihui Liu
- Neurointerventional Department, Huludao Central Hospital, Huludao, Liaoning, People's Republic of China
| | - Aiming Wang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Fan Zhang
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Zhiqiang Cui
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- School of Pharmacy, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jingwei Guan
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaohong Sun
- Department of Neurology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
- Science Experiment Center, China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
2
|
Huang Q, Tang J, Xiang Y, Shang X, Li K, Chen L, Hu J, Li H, Pi Y, Yang H, Zhang H, Tan H, Xiyang Y, Jin H, Li X, Chen M, Mao R, Wang Q. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione rescues oligodendrocytes ferroptosis leading to myelin loss and ameliorates neuronal injury facilitating memory in neonatal hypoxic-ischemic brain damage. Exp Neurol 2025; 390:115262. [PMID: 40246011 DOI: 10.1016/j.expneurol.2025.115262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Neonatal brain hypoxia-ischemia (HI) is proved to cause white matter injury (WMI), which resulted in behavioral disturbance. Myelin formed by oligodendrocytes vulnerable to hypoxia-ischemia (HI), regulating motor and cognitive function, is easily damaged by HI causing myelin loss. 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) has a potential rescue role in neuronal death post HI. Studies reported that neuronal ferroptosis could be induced by HI and linked to behavioral abnormalities. However, the effect of TDZD-8 on WMI and its involvement in memory recovery remains unclear. In this study, our HIBD model showed impaired memory function caused by neuronal injury and myelin loss. TDZD-8 effectively reversed this pathology. Underlying mechanistic exploration implied that TDZD-8 ameliorating myelin loss via ferroptosis pathway was involved in the process of TDZD-8 treating neonatal HIBD. In conclusion, our data demonstrated that combined effect of white matter repairment and neuronal protection achieved the therapeutic role of TDZD-8 in neonatal HIBD, and suggested that white matter repairment also could be a considerable clinical therapy for neonatal HIBD.
Collapse
Affiliation(s)
- Qiyi Huang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Jiahang Tang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - You Xiang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xinying Shang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Kunlin Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Lijia Chen
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Junnan Hu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Han Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanxiong Pi
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Haiyan Yang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Huijia Zhang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Heng Tan
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanbin Xiyang
- Institution of Neuroscience, Kunming Medical University, Kunming 650500, China
| | - Huiyan Jin
- Department of Functional Experiment, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xia Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Manjun Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Rongrong Mao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| | - Qian Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
3
|
Yamakawa A, Suganuma M, Mitsumori R, Niida S, Ozaki K, Shigemizu D. Alzheimer's disease may develop from changes in the immune system, cell cycle, and protein processing following alterations in ribosome function. Sci Rep 2025; 15:3838. [PMID: 39885278 PMCID: PMC11782650 DOI: 10.1038/s41598-025-88526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/29/2025] [Indexed: 02/01/2025] Open
Abstract
The prevalence of Alzheimer's disease (AD) is increasing as society ages. The details of AD pathogenesis have not been fully elucidated, and a comprehensive gene expression analysis of the process leading up to the onset of AD would be helpful for understanding the mechanism. We performed an RNA sequencing analysis on a cohort of 1227 Japanese blood samples, representing 424 AD patients, 543 individuals with mild cognitive impairment (MCI), and 260 cognitively normal (CN) individuals. A total of 883 and 1169 statistically significant differentially expressed genes (DEGs) were identified between CN and MCI (CN-MCI) and between MCI and AD (MCI-AD), respectively. Pathway analyses using these DEGs, followed by protein-protein interaction network analysis, revealed key roles of ribosomal function in MCI progression, whereas immune responses, cell cycle, and protein processing in endoplasmic reticulum were involved in AD progression. Our findings indicate that the onset of AD might be associated with gene expression changes in the immune system, cell cycle, and protein processing following alterations in the expression of ribosomal protein genes during the MCI stage, although validation using brain tissue samples will be necessary in the future. Given the known effectiveness of delaying MCI progression in preventing AD, the genes related to ribosomal function might emerge as biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Akiko Yamakawa
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Mutsumi Suganuma
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
| | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, 474-8511, Aichi, Japan
| | - Kouichi Ozaki
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8551, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Kanagawa, Japan
| | - Daichi Shigemizu
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, 474-8511, Aichi, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, 734-8551, Japan.
| |
Collapse
|
4
|
Dou RX, Zhang YM, Hu XJ, Gao FL, Zhang LL, Liang YH, Zhang YY, Yao YP, Yin L, Zhang Y, Gu C. Aβ 1-42 promotes microglial activation and apoptosis in the progression of AD by binding to TLR4. Redox Biol 2024; 78:103428. [PMID: 39550828 PMCID: PMC11615585 DOI: 10.1016/j.redox.2024.103428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024] Open
Abstract
Alzheimer's disease (AD) is one of the most common age-related neurodegenerative diseases and the most devastating form of senile dementia. It has a complex mechanism and no effective treatment. Exploring the pathogenesis of AD and providing ideas for treatment can effectively improve the prognosis of AD. Microglia were incubated with β-amyloid protein 1-42 (Aβ1-42) to construct an AD cell model. After microglia were activated, cell morphology changed, the expression level of inflammatory factors increased, cell apoptosis was promoted, and the expression of microtubule-associated protein (Tau protein) and related proteins increased. By up-regulating and down-regulating Toll-like receptor 4 (TLR4), the cells were divided into TLR4 knockdown negative control group(Lv-NC group), TLR4 knockdown group(Lv-TLR4 group), TLR4 overexpression negative control group(Sh-NC group), and TLR4 overexpression group(Sh-TLR4 group). The expression of inflammatory factors was detected again. It was found that compared with the Lv-NC group, the expression of various inflammatory factors in the Lv-TLR4 group decreased, cell apoptosis was inhibited, and the expression of Tau protein and related proteins decreased. Compared with the Sh-NC group, the expression of inflammatory factors in the Sh-TLR4 group increased, cell apoptosis was promoted, and the expression of Tau protein and related proteins increased. These results indicate that Aβ1-42 may promote microglial activation and apoptosis by binding to TLR4. Reducing the expression of TLR4 can reduce the occurrence of inflammatory response in AD cells and slow down cell apoptosis. Therefore, TLR4 is expected to become a new target for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Rui-Xia Dou
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China.
| | - Ya-Min Zhang
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Xiao-Juan Hu
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Fu-Lin Gao
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Lu-Lu Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yun-Hua Liang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yin-Ying Zhang
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China; Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Yu-Ping Yao
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Li Yin
- Western Medical District of Chinese PLA General Hospital, Haidian District, 100097, Beijing, China
| | - Yi Zhang
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China
| | - Cheng Gu
- Department of Neurology, Gansu Provincial People's Hospital, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
5
|
Zhang Y, Wang X, Huang J, Zhang X, Bu L, Zhang Y, Liang F, Wu S, Zhang M, Zhang L, Zhang L. CASIN exerts anti-aging effects through RPL4 on the skin of naturally aging mice. Aging Cell 2024; 23:e14333. [PMID: 39289787 PMCID: PMC11634736 DOI: 10.1111/acel.14333] [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: 03/25/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Skin aging has been associated with the onset of various skin issues, and recent studies have identified an increase in Cdc42 activity in naturally aging mice. While previous literature has suggested that CASIN, a specific inhibitor of Cdc42 activity, may possess anti-aging properties, its specific effects on the epidermis and dermis, as well as the underlying mechanisms in naturally aging mice, remain unclear. Our study revealed that CASIN demonstrated the ability to increase epidermal and dermal thickness, enhance dermal-epidermal junction, and stimulate collagen and elastic fiber synthesis in 9-, 15-, and 24-month-old C57BL/6 mice in vivo. Moreover, CASIN was found to enhance the proliferation, differentiation, and colony formation and restore the cytoskeletal morphology of primary keratinocytes in naturally aging skin in vitro. Furthermore, the anti-aging properties of CASIN on primary fibroblasts in aging mice were mediated by the ribosomal protein RPL4 using proteomic sequencing, influencing collagen synthesis and cytoskeletal morphology both in vitro and in vivo. Meanwhile, both subcutaneous injection and topical application exhibited anti-aging effects for a duration of 21 days. Additionally, CASIN exhibited anti-inflammatory properties, while reduced expression of RPL4 was associated with increased inflammation in the skin of naturally aging mice. Taken together, our results unveil a novel function of RPL4 in skin aging, providing a foundational basis for future investigations into ribosomal proteins. And CASIN shows promise as a potential anti-aging agent for naturally aging mouse skin, suggesting potential applications in the field.
Collapse
Affiliation(s)
- Yijia Zhang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Xueer Wang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Jianyuan Huang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Xinyue Zhang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Lingwei Bu
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Yarui Zhang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Fengting Liang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Shenhua Wu
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Min Zhang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Center for Orthopaedic Surgery of the Third Affiliated Hospital, Guangdong‐Hong Kong‐Macao Greater Bay Area Center for Brain Science and Brain‐Inspired IntelligenceSouthern Medical UniversityGuangzhouChina
| | - Lin Zhang
- GDMPA Key Laboratory of key Technologies for Cosmetics Safety and Efficacy Evaluation, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Histology and Embryology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
6
|
Malasala S, Azimian F, Chen YH, Twiss JL, Boykin C, Akhtar SN, Lu Q. Enabling systemic identification and functionality profiling for Cdc42 homeostatic modulators. Commun Chem 2024; 7:271. [PMID: 39562759 PMCID: PMC11577034 DOI: 10.1038/s42004-024-01352-7] [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: 05/25/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024] Open
Abstract
Maintaining body homeostasis is the ultimate key to health. There are rich resources of bioactive materials for the functionality of homeostatic modulators (HMs) from both natural and synthetic chemical repertories1-3. HMs are powerful modern therapeutics for human diseases including neuropsychiatric diseases, mental disorders, and drug addiction (e.g. Buspirone and benzodiazepines)4-7. However, the identification of therapeutic HMs are often unpredictable and limited to membrane protein receptors and ion channels. Based on a serendipitously encountered small molecule ZCL278 with partial agonist (PA) profile as a model compound8-10, the Mant-GTP fluorophore-based Cdc42-GEF (guanine nucleotide exchange factor) screening uncovered a near holistic spectrum of HMs for Cdc42, a cytoplasmic small GTPase in the Ras superfamily11,12. We categorized these HMs as functionally distinct, with some previously understudied classes: Class I-competitive PAs, Class II-hormetic agonists, Class III-bona fide inhibitors, Class IV-bona fide activators, and Class V-ligand-enhanced agonists. The model HMs elicited striking biological functionalities in modulating bradykinin activation of Cdc42 signaling as well as actin remodeling while they ameliorated Alzheimer's disease-like social behavior in mouse model. Furthermore, molecular structural modeling analyses led to the concept of preferential binding pocket order (PBPO) for profiling HMs that target Cdc42 complexed with intersectin (ITSN), a GEF selectively activating Cdc42. Remarkably, the PBPO enabled a prediction of HM class that mimics the pharmacological functionality. Therefore, our study highlights a model path to actively capture different classes of HM to broaden therapeutic landscape.
Collapse
Affiliation(s)
- Satyaveni Malasala
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Fereshteh Azimian
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Yan-Hua Chen
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery L Twiss
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
- Carolina Autism and Neurodevelopment Center, University of South Carolina, Columbia, SC, USA
| | - Christi Boykin
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
| | - Shayan Nik Akhtar
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA
| | - Qun Lu
- Department of Chemistry and Biochemistry, College of Arts and Sciences, University of South Carolina, Columbia, SC, USA.
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- The Harriet and John Wooten Laboratory for Alzheimer's and Neurodegenerative Diseases Research, The Brody School of Medicine, East Carolina University, Greenville, NC, USA.
- Laboratory of Molecular Neurotherapeutics, Center for Neurotherapeutics, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Malasala S, Azimian F, Chen YH, Twiss JL, Boykin C, Akhtar SN, Lu Q. Enabling Systemic Identification and Functionality Profiling for Cdc42 Homeostatic Modulators. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574351. [PMID: 38260445 PMCID: PMC10802479 DOI: 10.1101/2024.01.05.574351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Homeostatic modulation is pivotal in modern therapeutics. However, the discovery of bioactive materials to achieve this functionality is often random and unpredictive. Here, we enabled a systemic identification and functional classification of chemicals that elicit homeostatic modulation of signaling through Cdc42, a classical small GTPase of Ras superfamily. Rationally designed for high throughput screening, the capture of homeostatic modulators (HMs) along with molecular re-docking uncovered at least five functionally distinct classes of small molecules. This enabling led to partial agonists, hormetic agonists, bona fide activators and inhibitors, and ligand-enhanced agonists. Novel HMs exerted striking functionality in bradykinin-Cdc42 activation of actin remodelingand modified Alzheimer's disease-like behavior in mouse model. This concurrent computer-aided and experimentally empowered HM profiling highlights a model path for predicting HM landscape. One Sentence Summary With concurrent experimental biochemical profiling and in silico computer-aided drug discovery (CADD) analysis, this study enabled a systemic identification and holistic classification of Cdc42 homeostatic modulators (HMs) and demonstrated the power of CADD to predict HM classes that can mimic the pharmacological functionality of interests. Introduction Maintainingbody homeostasisis the ultimate keyto health. Thereare rich resources of bioactive materials for this functionality from both natural and synthetic chemical repertories including partial agonists (PAs) and various allosteric modulators. These homeostatic modulators (HMs) play a unique role in modern therapeutics for human diseases such as mental disorders and drug addiction. Buspirone, for example, acts as a PA for serotonin 5-HT 1A receptor but is an antagonist of the dopamine D 2 receptor. Such medical useto treat general anxietydisorders (GADs) has become one of the most-commonly prescribed medications. However, most HMs in current uses target membrane proteins and are often derived from random discoveries. HMs as therapeutics targeting cytoplasmic proteins are even more rare despite that they are in paramount needs (e. g. targeting Ras superfamily small GTPases). Rationale Cdc42, a classical member of small GTPases of Ras superfamily, regulates PI3K-AKT and Raf-MEK-ERK pathways and has been implicated in various neuropsychiatric and mental disorders as well as addictive diseases and cancer. We previously reported the high-throughput in-silico screening followed by biological characterization of novel small molecule modulators (SMMs) of Cdc42-intersectin (ITSN) protein-protein interactions (PPIs). Based on a serendipitously discovered SMM ZCL278 with PA profile as a model compound, we hypothesized that there are more varieties of such HMs of Cdc42 signaling, and the model HMs can be defined by their distinct Cdc42-ITSN binding mechanisms using computer-aided drug discovery (CADD) analysis. We further reasoned that molecular modeling coupled with experimental profiling can predict HM spectrum and thus open the door for the holistic identification and classification of multifunctional cytoplasmic target-dependent HMs as therapeutics. Results The originally discovered Cdc42 inhibitor ZCL278 displaying PA properties prompted the inquiry whether this finding represented a random encounter of PAs or whether biologically significant PAs can be widely present. The top ranked compounds were initially defined by structural fitness and binding scores to Cdc42. Because higher binding scores do not necessarily translate to higher functionality, we performed exhaustive experimentations with over 2,500 independent Cdc42-GEF (guanine nucleotide exchange factor) assays to profile the GTP loading activities on all 44 top ranked compounds derived from the SMM library. The N-MAR-GTP fluorophore-based Cdc42-GEF assay platform provided the first glimpse of the breadth of HMs. A spectrum of Cdc42 HMs was uncovered that can be categorized into five functionally distinct classes: Class I-partial competitive agonists, Class II-hormetic agonists, Class III- bona fide inhibitors (or inverse agonists), Class IV- bona fide activators or agonists, and Class V-ligand-enhanced agonists. Remarkably, model HMs such as ZCL278, ZCL279, and ZCL367 elicited striking biological functionality in bradykinin-Cdc42 activation of actin remodeling and modified Alzheimer's disease (AD)-like behavior in mouse model. Concurrently, we applied Schrödinger-enabled analyses to perform CADD predicted classification of Cdc42 HMs. We modified the classic molecular docking to instill a preferential binding pocket order (PBPO) of Cdc42-ITSN, which was based on the five binding pockets in interface of Cdc42-ITSN. We additionally applied a structure-based pharmacophore hypothesis generation for the model compounds. Then, using Schrödinger's Phase Shape, 3D ligand alignments assigned HMs to Class I, II, III, IV, and V compounds. In this HM library compounds, PBPO, matching pharmacophoric featuring, and shape alignment, all put ZCL993 in Class II compound category, which was confirmed in the Cdc42-GEF assay. Conclusion HMs can target diseased cells or tissues while minimizing impacts on tissues that are unaffected. Using Cdc42 HM model compounds as a steppingstone, GTPase activation-based screening of SMM library uncovered five functionally distinct Cdc42 HM classes among which novel efficacies towards alleviating dysregulated AD-like features in mice were identified. Furthermore, molecular re-docking of HM model compounds led to the concept of PBPO. The CADD analysis with PBPO revealed similar profile in a color-coded spectrum to these five distinct classes of Cdc42 HMs identified by biochemical functionality-based screening. The current study enabled a systemic identification and holistic classification of Cdc42 HMs and demonstrated the power of CADD to predict an HM category that can mimic the pharmacological functionality of interests. With artificial intelligence/machine learning (AI/ML) on the horizon to mirror experimental pharmacological discovery like AlphaFold for protein structure prediction, our study highlights a model path to actively capture and profile HMs in potentially any PPI landscape. Graphic Abstract Identification and functional classification of Cdc42 homeostatic modulators HMs Using Cdc42 HM model compounds as reference, GTPase activation-based screening of compound libraries uncovered five functionally distinct Cdc42 HM classes. HMs showed novel efficacies towards alleviating dysregulated Alzheimer's disease (AD)-like behavioral and molecular deficits. In parallel, molecular re-docking of HM model compounds established their preferential binding pocket orders (PBPO). PBPO-based profiling (Red reflects the most, whereas green reflects the least, preferable binding pocket) revealed trends of similar pattern to the five classes from the functionality-based classification.
Collapse
|
10
|
Young JJ, Park HJ, Kim M, Par-Young J, Bartlett H, Kim HS, Unlu S, Osmani L, Shin MS, Bucala R, van Dyck CH, Allore H, Mecca AP, You S, Kang I. Aging gene signature of memory CD8 + T cells is associated with neurocognitive functioning in Alzheimer's disease. Immun Ageing 2023; 20:71. [PMID: 38042785 PMCID: PMC10693128 DOI: 10.1186/s12979-023-00396-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND Memory CD8+ T cells expand with age. We previously demonstrated an age-associated expansion of effector memory (EM) CD8+ T cells expressing low levels of IL-7 receptor alpha (IL-7Rαlow) and the presence of its gene signature (i.e., IL-7Rαlow aging genes) in peripheral blood of older adults without Alzheimer's disease (AD). Considering age as the strongest risk factor for AD and the recent finding of EM CD8+ T cell expansion, mostly IL-7Rαlow cells, in AD, we investigated whether subjects with AD have alterations in IL-7Rαlow aging gene signature, especially in relation to genes possibly associated with AD and disease severity. RESULTS We identified a set of 29 candidate genes (i.e., putative AD genes) which could be differentially expressed in peripheral blood of patients with AD through the systematic search of publicly available datasets. Of the 29 putative AD genes, 9 genes (31%) were IL-7Rαlow aging genes (P < 0.001), suggesting the possible implication of IL-7Rαlow aging genes in AD. These findings were validated by RT-qPCR analysis of 40 genes, including 29 putative AD genes, additional 9 top IL-7R⍺low aging but not the putative AD genes, and 2 inflammatory control genes in peripheral blood of cognitively normal persons (CN, 38 subjects) and patients with AD (40 mild cognitive impairment and 43 dementia subjects). The RT-qPCR results showed 8 differentially expressed genes between AD and CN groups; five (62.5%) of which were top IL-7Rαlow aging genes (FGFBP2, GZMH, NUAK1, PRSS23, TGFBR3) not previously reported to be altered in AD. Unbiased clustering analysis revealed 3 clusters of dementia patients with distinct expression levels of the 40 analyzed genes, including IL-7Rαlow aging genes, which were associated with neurocognitive function as determined by MoCA, CDRsob and neuropsychological testing. CONCLUSIONS We report differential expression of "normal" aging genes associated with IL-7Rαlow EM CD8+ T cells in peripheral blood of patients with AD, and the significance of such gene expression in clustering subjects with dementia due to AD into groups with different levels of cognitive functioning. These results provide a platform for studies investigating the possible implications of age-related immune changes, including those associated with CD8+ T cells, in AD.
Collapse
Affiliation(s)
- Juan Joseph Young
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Hong-Jai Park
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Minhyung Kim
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennefer Par-Young
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Hugh Bartlett
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
- Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Hye Sun Kim
- Yale School of Public Health, New Haven, CT, USA
| | - Serhan Unlu
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
- Cleveland Clinic Fairview Hospital, Cleveland, OH, USA
| | - Lais Osmani
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Min Sun Shin
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Richard Bucala
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Christopher H van Dyck
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Heather Allore
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
- Yale School of Public Health, New Haven, CT, USA
| | - Adam P Mecca
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Sungyong You
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Insoo Kang
- Department of Psychiatry, Yale School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|