1
|
Chen X, Wang YJ, Mu TW. Proteostasis regulation of GABA A receptors in neuronal function and disease. Biomed Pharmacother 2025; 186:117992. [PMID: 40112516 PMCID: PMC12068001 DOI: 10.1016/j.biopha.2025.117992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
The γ-aminobutyric acid type A receptors (GABAARs) are ligand-gated anion channels that mediate fast inhibitory neurotransmission in the mammalian central nervous system. GABAARs form heteropentameric assemblies comprising two α1, two β2, and one γ2 subunits as the most common subtype in mammalian brains. Proteostasis regulation of GABAARs involves subunit folding within the endoplasmic reticulum, assembling into heteropentamers, receptor trafficking to the cell surface, and degradation of terminally misfolded subunits. As GABAARs are surface proteins, their trafficking to the plasma membrane is critical for proper receptor function. Thus, variants in the genes encoding GABAARs that disrupt proteostasis result in various neurodevelopmental disorders, ranging from intellectual disability to idiopathic generalized epilepsy. This review summarizes recent progress about how the proteostasis network regulates protein folding, assembly, degradation, trafficking, and synaptic clustering of GABAARs. Additionally, emerging pharmacological approaches that restore proteostasis of pathogenic GABAAR variants are presented, providing a promising strategy to treat related neurological diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
2
|
Doerksen AH, Herath NN, Sanders SS. Fat traffic control: S-acylation in axonal transport. Mol Pharmacol 2025; 107:100039. [PMID: 40349611 DOI: 10.1016/j.molpha.2025.100039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Neuronal axons serve as a conduit for the coordinated transport of essential molecular cargo between structurally and functionally distinct subcellular compartments via axonal molecular machinery. Long-distance, efficient axonal transport of membrane-bound organelles enables signal transduction and neuronal homeostasis. Efficient axonal transport is conducted by dynein and kinesin ATPase motors that use a local ATP supply from metabolic enzymes tethered to transport vesicles. Molecular motor adaptor proteins promote the processive motility and cargo selectivity of fast axonal transport. Axonal transport impairments are directly causative or associated with many neurodegenerative diseases and neuropathologies. Cargo specificity, cargo-adaptor proteins, and posttranslational modifications of cargo, adaptor proteins, microtubules, or the motor protein subunits all contribute to the precise regulation of vesicular transit. One posttranslational lipid modification that is particularly important in neurons in regulating protein trafficking, protein-protein interactions, and protein association with lipid membranes is S-acylation. Interestingly, many fast axonal transport cargos, cytoskeletal-associated proteins, motor protein subunits, and adaptors are S-acylated to modulate axonal transport. Here, we review the established regulatory role of S-acylation in fast axonal transport and provide evidence for a broader role of S-acylation in regulating the motor-cargo complex machinery, adaptor proteins, and metabolic enzymes from low-throughput studies and S-acyl-proteomic data sets. We propose that S-acylation regulates fast axonal transport and vesicular motility through localization of the proteins required for the motile cargo-complex machinery and relate how perturbed S-acylation contributes to transport impairments in neurological disorders. SIGNIFICANCE STATEMENT: This review investigates the regulatory role of S-acylation in fast axonal transport and its connection to neurological diseases, with a focus on the emerging connections between S-acylation and the molecular motors, adaptor proteins, and metabolic enzymes that make up the trafficking machinery.
Collapse
Affiliation(s)
- Amelia H Doerksen
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Ontario, Canada
| | - Nisandi N Herath
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Ontario, Canada
| | - Shaun S Sanders
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Ontario, Canada.
| |
Collapse
|
3
|
Welle TM, Smith KR. Release your inhibitions: The cell biology of GABAergic postsynaptic plasticity. Curr Opin Neurobiol 2025; 90:102952. [PMID: 39721557 PMCID: PMC11839402 DOI: 10.1016/j.conb.2024.102952] [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: 09/20/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
GABAergic synaptic inhibition controls circuit function by regulating neuronal plasticity, excitability, and firing. To achieve these goals, inhibitory synapses themselves undergo several forms of plasticity via diverse mechanisms, strengthening and weakening phasic inhibition in response to numerous activity-induced stimuli. These mechanisms include changing the number and arrangement of functional GABAARs within the inhibitory postsynaptic domain (iPSD), which can profoundly regulate inhibitory synapse strength. Here, we explore recent advances in our molecular understanding of inhibitory postsynaptic plasticity, with a focus on modulation of the trafficking, protein-protein interactions, nanoscale-organization, and posttranscriptional regulation of GABAARs and iPSD proteins. What has emerged is a complex mechanistic picture of how synaptic inhibition is controlled, with critical ramifications for cognition under typical and pathogenic conditions.
Collapse
Affiliation(s)
- Theresa M Welle
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
4
|
Teoh J, Bartolini F. Emerging roles for tubulin PTMs in neuronal function and neurodegenerative disease. Curr Opin Neurobiol 2025; 90:102971. [PMID: 39862522 PMCID: PMC11839326 DOI: 10.1016/j.conb.2025.102971] [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: 08/22/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Neurons are equipped with microtubules of different stability with stable and dynamic domains often coexisting on the same microtubule. While dynamic microtubules undergo random transitions between disassembly and assembly, stable ones persist long enough to serve as platforms for tubulin-modifying enzymes (known as writers) that attach molecular components to the α- or β-tubulin subunits. The combination of these posttranslational modifications (PTMs) results in a "tubulin code," dictating the behavior of selected proteins (known as readers), some of which were shown to be crucial for neuronal function. Recent research has further highlighted that disturbances in tubulin PTMs can lead to neurodegeneration, sparking an emerging field of investigation with numerous questions such as whether and how tubulin PTMs can affect neurotransmission and synaptic plasticity and whether restoring balanced tubulin PTM levels could effectively prevent or mitigate neurodegenerative disease.
Collapse
Affiliation(s)
- JiaJie Teoh
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA
| | - Francesca Bartolini
- Department of Pathology & Cell Biology, Columbia University Irving Medical Center, 10032, New York, NY, USA.
| |
Collapse
|
5
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. GeroScience 2024; 46:5819-5841. [PMID: 38509416 PMCID: PMC11493911 DOI: 10.1007/s11357-024-01133-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: 01/08/2024] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent subcutaneous bioidentical E2 chronic treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p = 1.6 × 10-51) and upregulation (p = 3.8 × 10-3) of UBE2M across both brain regions provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p = 1.9 × 10-4; interaction p = 3.5 × 10-2) of LTBR in the PFC provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step toward understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Kip D Zimmerman
- Center for Precision Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Larry Wilhelm
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Dongqin Zhu
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Jessica Bodie
- Department of Translational Neuroscience, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Henryk F Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| |
Collapse
|
6
|
Ji X, Liu S, Li S, Li X, Luo A, Zhang X, Zhao Y. GABA in early brain development: A dual role review. Int J Dev Neurosci 2024; 84:843-856. [PMID: 39503050 DOI: 10.1002/jdn.10387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 01/03/2025] Open
Abstract
This comprehensive review examines the multifaceted roles of gamma-aminobutyric acid (GABA) in early brain development. GABA, traditionally recognized for its inhibitory functions in the mature brain, also exhibits excitatory effects during early neural development. This article explores the mechanisms behind GABA's dual roles, detailing its impact on the properties of the immature brain, the mechanisms of GABA-mediated excitation, the role of GABA-mediated presynaptic inhibition, the trophic actions of GABA during early development, GABA regulation of neurite growth and GABA-mediated cell differentiation in the immature brain. Emphasizing recent research findings, the review highlights the significance of GABAergic signalling in shaping the developing brain and its potential implications for understanding neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaoyu Ji
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuzhen Liu
- Department of Anesthesiology, Tai'an Central Hospital, Tai'an, China
| | - Shiyong Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Zhao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Wu Y, Wang Y, Lu Y, Yan J, Zhao H, Yang R, Pan J. Research advances in huntingtin-associated protein 1 and its application prospects in diseases. Front Neurosci 2024; 18:1402996. [PMID: 38975245 PMCID: PMC11224548 DOI: 10.3389/fnins.2024.1402996] [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: 03/18/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Huntingtin-associated protein 1 (HAP1) was the first protein discovered to interact with huntingtin. Besides brain, HAP1 is also expressed in the spinal cord, dorsal root ganglion, endocrine, and digestive systems. HAP1 has diverse functions involving in vesicular transport, receptor recycling, gene transcription, and signal transduction. HAP1 is strongly linked to several neurological diseases, including Huntington's disease, Alzheimer's disease, epilepsy, ischemic stroke, and depression. In addition, HAP1 has been proved to participate in cancers and diabetes mellitus. This article provides an overview of HAP1 regarding the tissue distribution, cell localization, functions, and offers fresh perspectives to investigate its role in diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
8
|
Banerjee S, Zhao Q, Wang B, Qin J, Yuan X, Lou Z, Zheng W, Li H, Wang X, Cheng X, Zhu Y, Lin F, Yang F, Xu J, Munshi A, Das P, Zhou Y, Mandal K, Wang Y, Ayub M, Hirokawa N, Xi Y, Chen G, Li C. A novel in-frame deletion in KIF5C gene causes infantile onset epilepsy and psychomotor retardation. MedComm (Beijing) 2024; 5:e469. [PMID: 38525108 PMCID: PMC10960728 DOI: 10.1002/mco2.469] [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: 05/26/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 03/26/2024] Open
Abstract
Motor proteins, encoded by Kinesin superfamily (KIF) genes, are critical for brain development and plasticity. Increasing studies reported KIF's roles in neurodevelopmental disorders. Here, a 6 years and 3 months-old Chinese boy with markedly symptomatic epilepsy, intellectual disability, brain atrophy, and psychomotor retardation was investigated. His parents and younger sister were phenotypically normal and had no disease-related family history. Whole exome sequencing identified a novel heterozygous in-frame deletion (c.265_267delTCA) in exon 3 of the KIF5C in the proband, resulting in the removal of evolutionarily highly conserved p.Ser90, located in its ATP-binding domain. Sanger sequencing excluded the proband's parents and family members from harboring this variant. The activity of ATP hydrolysis in vitro was significantly reduced as predicted. Immunofluorescence studies showed wild-type KIF5C was widely distributed throughout the cytoplasm, while mutant KIF5C was colocalized with microtubules. The live-cell imaging of the cargo-trafficking assay revealed that mutant KIF5C lost the peroxisome-transporting ability. Drosophila models also confirmed p.Ser90del's essential role in nervous system development. This study emphasized the importance of the KIF5C gene in intracellular cargo-transport as well as germline variants that lead to neurodevelopmental disorders and might enable clinicians for timely and accurate diagnosis and disease management in the future.
Collapse
Affiliation(s)
- Santasree Banerjee
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
- Department of GeneticsCollege of Basic Medical SciencesJilin UniversityChangchunChina
- Department of GeneticsUniversity of DelhiNew DelhiIndia
| | - Qiang Zhao
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Bo Wang
- Department of PediatricsShenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Jiale Qin
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Xin Yuan
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Ziwei Lou
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Weizeng Zheng
- Department of RadiologyWomen's HospitalZhejiang University School of MedicineHangzhouChina
| | - Huanguo Li
- Department of RadiologyHangzhou Hospital of Traditional Chinese MedicineHangzhouChina
| | - Xiaojun Wang
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Xiawei Cheng
- School of PharmacyEast China University of Science and TechnologyShanghaiChina
| | - Yu Zhu
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Fan Lin
- Department of Cell BiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingChina
| | - Fan Yang
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Junyu Xu
- Department of Neurobiology, Department of Rehabilitation and Department of Internal Medicine of the Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Anjana Munshi
- Department of Human Genetics and Molecular MedicineCentral University of PunjabBathindaIndia
| | - Parimal Das
- Centre for Genetic DisordersBanaras Hindu UniversityVaranasiIndia
| | - Yuanfeng Zhou
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Kausik Mandal
- Department of Medical GeneticsSanjay Gandhi Postgraduate Institute of Medical SciencesLucknowUttar PradeshIndia
| | - Yi Wang
- Department of Neurology and Epilepsy CenterChildren's Hospital of Fudan UniversityShanghaiChina
| | - Muhammad Ayub
- Department of PsychiatryUniversity College LondonLondonUK
| | - Nobutaka Hirokawa
- Department of Cell Biology and AnatomyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Yongmei Xi
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
| | - Guangfu Chen
- Department of PediatricsShenzhen Second People's HospitalThe First Affiliated Hospital of Shenzhen University Health Science CenterShenzhenChina
| | - Chen Li
- Department of Human Genetics and Department of Ultrasound, Women's HospitalSchool of Basic Medical ScienceZhejiang Provincial Key Laboratory of Genetic and Developmental DisordersZhejiang University School of MedicineHangzhouChina
- Alibaba‐Zhejiang University Joint Research Center of Future Digital HealthcareHangzhouChina
| |
Collapse
|
9
|
Xie MX, Lai RC, Xiao YB, Zhang X, Cao XY, Tian XY, Chen AN, Chen ZY, Cao Y, Li X, Zhang XL. Endophilin A2 controls touch and mechanical allodynia via kinesin-mediated Piezo2 trafficking. Mil Med Res 2024; 11:17. [PMID: 38475827 DOI: 10.1186/s40779-024-00520-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 02/02/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Tactile and mechanical pain are crucial to our interaction with the environment, yet the underpinning molecular mechanism is still elusive. Endophilin A2 (EndoA2) is an evolutionarily conserved protein that is documented in the endocytosis pathway. However, the role of EndoA2 in the regulation of mechanical sensitivity and its underlying mechanisms are currently unclear. METHODS Male and female C57BL/6 mice (8-12 weeks) and male cynomolgus monkeys (7-10 years old) were used in our experiments. Nerve injury-, inflammatory-, and chemotherapy-induced pathological pain models were established for this study. Behavioral tests of touch, mechanical pain, heat pain, and cold pain were performed in mice and nonhuman primates. Western blotting, immunostaining, co-immunoprecipitation, proximity ligation and patch-clamp recordings were performed to gain insight into the mechanisms. RESULTS The results showed that EndoA2 was primarily distributed in neurofilament-200-positive (NF200+) medium-to-large diameter dorsal root ganglion (DRG) neurons of mice and humans. Loss of EndoA2 in mouse NF200+ DRG neurons selectively impaired the tactile and mechanical allodynia. Furthermore, EndoA2 interacted with the mechanically sensitive ion channel Piezo2 and promoted the membrane trafficking of Piezo2 in DRG neurons. Moreover, as an adaptor protein, EndoA2 also bound to kinesin family member 5B (KIF5B), which was involved in the EndoA2-mediated membrane trafficking process of Piezo2. Loss of EndoA2 in mouse DRG neurons damaged Piezo2-mediated rapidly adapting mechanically activated currents, and re-expression of EndoA2 rescued the MA currents. In addition, interference with EndoA2 also suppressed touch sensitivity and mechanical hypersensitivity in nonhuman primates. CONCLUSIONS Our data reveal that the KIF5B/EndoA2/Piezo2 complex is essential for Piezo2 trafficking and for sustaining transmission of touch and mechanical hypersensitivity signals. EndoA2 regulates touch and mechanical allodynia via kinesin-mediated Piezo2 trafficking in sensory neurons. Our findings identify a potential new target for the treatment of mechanical pain.
Collapse
Affiliation(s)
- Man-Xiu Xie
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Ren-Chun Lai
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yi-Bin Xiao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xi Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xian-Ying Cao
- Engineering Technology Research Center for Elderly Health Management in Hainan Province, Haikou, 571137, China
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao-Yu Tian
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - An-Nan Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zi-Yi Chen
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Cao
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao Li
- College of Food Science and Technology, Hainan University, Haikou, 570228, China
| | - Xiao-Long Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
10
|
Abdi G, Jain M, Patil N, Upadhyay B, Vyas N, Dwivedi M, Kaushal RS. 14-3-3 proteins-a moonlight protein complex with therapeutic potential in neurological disorder: in-depth review with Alzheimer's disease. Front Mol Biosci 2024; 11:1286536. [PMID: 38375509 PMCID: PMC10876095 DOI: 10.3389/fmolb.2024.1286536] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/05/2024] [Indexed: 02/21/2024] Open
Abstract
Alzheimer's disease (AD) affects millions of people worldwide and is a gradually worsening neurodegenerative condition. The accumulation of abnormal proteins, such as tau and beta-amyloid, in the brain is a hallmark of AD pathology. 14-3-3 proteins have been implicated in AD pathology in several ways. One proposed mechanism is that 14-3-3 proteins interact with tau protein and modulate its phosphorylation, aggregation, and toxicity. Tau is a protein associated with microtubules, playing a role in maintaining the structural integrity of neuronal cytoskeleton. However, in the context of Alzheimer's disease (AD), an abnormal increase in its phosphorylation occurs. This leads to the aggregation of tau into neurofibrillary tangles, which is a distinctive feature of this condition. Studies have shown that 14-3-3 proteins can bind to phosphorylated tau and regulate its function and stability. In addition, 14-3-3 proteins have been shown to interact with beta-amyloid (Aβ), the primary component of amyloid plaques in AD. 14-3-3 proteins can regulate the clearance of Aβ through the lysosomal degradation pathway by interacting with the lysosomal membrane protein LAMP2A. Dysfunction of lysosomal degradation pathway is thought to contribute to the accumulation of Aβ in the brain and the progression of AD. Furthermore, 14-3-3 proteins have been found to be downregulated in the brains of AD patients, suggesting that their dysregulation may contribute to AD pathology. For example, decreased levels of 14-3-3 proteins in cerebrospinal fluid have been suggested as a biomarker for AD. Overall, these findings suggest that 14-3-3 proteins may play an important role in AD pathology and may represent a potential therapeutic target for the disease. However, further research is needed to fully understand the mechanisms underlying the involvement of 14-3-3 proteins in AD and to explore their potential as a therapeutic target.
Collapse
Affiliation(s)
- Gholamareza Abdi
- Department of Biotechnology, Persian Gulf Research Institute, Persian Gulf University, Bushehr, Iran
| | - Mukul Jain
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Nil Patil
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Bindiya Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
| | - Nigam Vyas
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Biophysics and Structural Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
| | - Manish Dwivedi
- Amity Institute of Biotechnology, Amity University, Lucknow, Uttar Pradesh, India
| | - Radhey Shyam Kaushal
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Biophysics and Structural Biology Laboratory, Research and Development Cell, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
11
|
Cervera-Juanes R, Zimmerman KD, Wilhelm L, Zhu D, Bodie J, Kohama SG, Urbanski HF. Modulation of neural gene networks by estradiol in old rhesus macaque females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572105. [PMID: 38187564 PMCID: PMC10769303 DOI: 10.1101/2023.12.18.572105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The postmenopausal decrease in circulating estradiol (E2) levels has been shown to contribute to several adverse physiological and psychiatric effects. To elucidate the molecular effects of E2 on the brain, we examined differential gene expression and DNA methylation (DNAm) patterns in the nonhuman primate brain following ovariectomy (Ov) and subsequent E2 treatment. We identified several dysregulated molecular networks, including MAPK signaling and dopaminergic synapse response, that are associated with ovariectomy and shared across two different brain areas, the occipital cortex (OC) and prefrontal cortex (PFC). The finding that hypomethylation (p=1.6×10-51) and upregulation (p=3.8×10-3) of UBE2M across both brain regions, provide strong evidence for molecular differences in the brain induced by E2 depletion. Additionally, differential expression (p=1.9×10-4; interaction p=3.5×10-2) of LTBR in the PFC, provides further support for the role E2 plays in the brain, by demonstrating that the regulation of some genes that are altered by ovariectomy may also be modulated by Ov followed by hormone replacement therapy (HRT). These results present real opportunities to understand the specific biological mechanisms that are altered with depleted E2. Given E2's potential role in cognitive decline and neuroinflammation, our findings could lead to the discovery of novel therapeutics to slow cognitive decline. Together, this work represents a major step towards understanding molecular changes in the brain that are caused by ovariectomy and how E2 treatment may revert or protect against the negative neuro-related consequences caused by a depletion in estrogen as women approach menopause.
Collapse
Affiliation(s)
- Rita Cervera-Juanes
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Kip D. Zimmerman
- Center for Precision Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
- Department of Internal Medicine, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Larry Wilhelm
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Dongqin Zhu
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Jessica Bodie
- Department of Translational Neuroscience, Atrium Health Wake Forest Baptist, Winston-Salem, NC 27157
| | - Steven G. Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
12
|
Makeeva VS, Dyrkheeva NS, Lavrik OI, Zakian SM, Malakhova AA. Mutant-Huntingtin Molecular Pathways Elucidate New Targets for Drug Repurposing. Int J Mol Sci 2023; 24:16798. [PMID: 38069121 PMCID: PMC10706709 DOI: 10.3390/ijms242316798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The spectrum of neurodegenerative diseases known today is quite extensive. The complexities of their research and treatment lie not only in their diversity. Even many years of struggle and narrowly focused research on common pathologies such as Alzheimer's, Parkinson's, and other brain diseases have not brought cures for these illnesses. What can be said about orphan diseases? In particular, Huntington's disease (HD), despite affecting a smaller part of the human population, still attracts many researchers. This disorder is known to result from a mutation in the HTT gene, but having this information still does not simplify the task of drug development and studying the mechanisms of disease progression. Nonetheless, the data accumulated over the years and their analysis provide a good basis for further research. Here, we review studies devoted to understanding the mechanisms of HD. We analyze genes and molecular pathways involved in HD pathogenesis to describe the action of repurposed drugs and try to find new therapeutic targets.
Collapse
Affiliation(s)
- Vladlena S. Makeeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia;
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia;
| | - Suren M. Zakian
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| | - Anastasia A. Malakhova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| |
Collapse
|
13
|
Garcia JD, Wolfe SE, Stewart AR, Tiemeier E, Gookin SE, Guerrero MB, Quillinan N, Smith KR. Distinct mechanisms drive sequential internalization and degradation of GABA ARs during global ischemia and reperfusion injury. iScience 2023; 26:108061. [PMID: 37860758 PMCID: PMC10582478 DOI: 10.1016/j.isci.2023.108061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/30/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Synaptic inhibition is critical for controlling neuronal excitability and function. During global cerebral ischemia (GCI), inhibitory synapses are rapidly eliminated, causing hyper-excitability which contributes to cell-death and the pathophysiology of disease. Sequential disassembly of inhibitory synapses begins within minutes of ischemia onset: GABAARs are rapidly trafficked away from the synapse, the gephyrin scaffold is removed, followed by loss of the presynaptic terminal. GABAARs are endocytosed during GCI, but how this process accompanies synapse disassembly remains unclear. Here, we define the precise trafficking itinerary of GABAARs during the initial stages of GCI, placing them in the context of rapid synapse elimination. Ischemia-induced GABAAR internalization quickly follows their initial dispersal from the synapse, and is controlled by PP1α signaling. During reperfusion injury, GABAARs are then trafficked to lysosomes for degradation, leading to permanent removal of synaptic GABAARs and contributing to the profound reduction in synaptic inhibition observed hours following ischemia onset.
Collapse
Affiliation(s)
- Joshua D. Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Sarah E. Wolfe
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Amber R. Stewart
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Erika Tiemeier
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Sara E. Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Mayra Bueno Guerrero
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Katharine R. Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
14
|
Beatriz M, Rodrigues RJ, Vilaça R, Egas C, Pinheiro PS, Daley GQ, Schlaeger TM, Raimundo N, Rego AC, Lopes C. Extracellular vesicles improve GABAergic transmission in Huntington's disease iPSC-derived neurons. Theranostics 2023; 13:3707-3724. [PMID: 37441602 PMCID: PMC10334823 DOI: 10.7150/thno.81981] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/22/2023] [Indexed: 07/15/2023] Open
Abstract
Background: Extracellular vesicles (EVs) carry bioactive molecules associated with various biological processes, including miRNAs. In both Huntington's disease (HD) models and human samples, altered expression of miRNAs involved in synapse regulation was reported. Recently, the use of EV cargo to reverse phenotypic alterations in disease models with synaptopathy as the end result of the pathophysiological cascade has become an interesting possibility. Methods: Here, we assessed the contribution of EVs to GABAergic synaptic alterations using a human HD model and studied the miRNA content of isolated EVs. Results: After differentiating human induced pluripotent stem cells into electrophysiologically active striatal-like GABAergic neurons, we found that HD-derived neurons displayed reduced density of inhibitory synapse markers and GABA receptor-mediated ionotropic signaling. Treatment with EVs secreted by control (CTR) fibroblasts reversed the deficits in GABAergic synaptic transmission and increased the density of inhibitory synapses in HD-derived neuron cultures, while EVs from HD-derived fibroblasts had the opposite effects on CTR-derived neurons. Moreover, analysis of miRNAs from purified EVs identified a set of differentially expressed miRNAs between manifest HD, premanifest, and CTR lines with predicted synaptic targets. Conclusion: The EV-mediated reversal of the abnormal GABAergic phenotype in HD-derived neurons reinforces the potential role of EV-miRNAs on synapse regulation.
Collapse
Affiliation(s)
- Margarida Beatriz
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Ricardo J. Rodrigues
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rita Vilaça
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Conceição Egas
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- Biocant- Transfer Technology Association, Biocant Park, Cantanhede, Portugal
| | - Paulo S. Pinheiro
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Sciences and Technology, Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - George Q. Daley
- Division of Pediatric Hematology/Oncology, Children's Hospital Boston, Boston, MA USA
- Harvard Stem Cell Institute, Boston, MA USA
| | - Thorsten M. Schlaeger
- Division of Pediatric Hematology/Oncology, Children's Hospital Boston, Boston, MA USA
- Harvard Stem Cell Institute, Boston, MA USA
| | - Nuno Raimundo
- MIA - Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - A. Cristina Rego
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- FMUC - Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Carla Lopes
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- MIA - Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Abstract
Neurons are markedly compartmentalized, which makes them reliant on axonal transport to maintain their health. Axonal transport is important for anterograde delivery of newly synthesized macromolecules and organelles from the cell body to the synapse and for the retrograde delivery of signaling endosomes and autophagosomes for degradation. Dysregulation of axonal transport occurs early in neurodegenerative diseases and plays a key role in axonal degeneration. Here, we provide an overview of mechanisms for regulation of axonal transport; discuss how these mechanisms are disrupted in neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, hereditary spastic paraplegia, amyotrophic lateral sclerosis, and Charcot-Marie-Tooth disease; and discuss therapeutic approaches targeting axonal transport.
Collapse
|
16
|
Yin JB, Liu HX, Dong QQ, Wu HH, Liang ZW, Fu JT, Zhao WJ, Hu HQ, Guo HW, Zhang T, Lu YC, Jin S, Wang XL, Cao BZ, Wang Z, Ding T. Correlative increasing expressions of KIF5b and Nav1.7 in DRG neurons of rats under neuropathic pain conditions. Physiol Behav 2023; 263:114115. [PMID: 36773735 DOI: 10.1016/j.physbeh.2023.114115] [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: 09/11/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Nav1.7, one of tetrodotoxin-sensitive voltage-gated sodium channels, mainly expressed in the small diameter dorsal root ganglion (DRG) neurons. The expression and accumulation on neuronal membrane of Nav1.7 increased following peripheral tissue inflammation or nerve injury. However, the mechanisms for membrane accumulation of Nav1.7 remained unclear. We report that KIF5b, a highly expressed member of the kinesin-1 family in DRGs, promoted the translocation of Nav1.7 to the plasma membrane in DRG neurons of the rat. Following nociceptive behaviors in rats induced by peripheral spared nerve injury (SNI), synchronously increased KIF5b and Nav1.7 expressions were observed in DRGs. Immunohistochemistry staining demonstrated the co-expressions of KIF5b and Nav1.7 in the same DRG neurons. Immunoprecipitation experiments further confirmed the interactions between KIF5b and Nav1.7. Moreover, intrathecal injections of KIF5b shRNA moderated the SNI-induced both mechanical and thermal hyperalgesia. The rescued analgesic effects also alleviated SNI-induced anxiety-like behaviors. In sum, KIF5b was required for the membrane localizations of Nav1.7, which suggests a novel mechanism for the trafficking of Nav1.7 involved in neuropathic pain.
Collapse
Affiliation(s)
- Jun-Bin Yin
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China; Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Hai-Xia Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Jinan 250021, China
| | - Qin-Qin Dong
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China; Department of Neurology, Jinzhou Medical University, Jinzhou 121000, China
| | - Huang-Hui Wu
- Department of Anesthesiology, Medical College of Xiamen University, Xiamen 361005, China
| | - Zhuo-Wen Liang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Jin-Tao Fu
- Department of Critical Care Medicine, Affiliated Yanzhou District Hospital of Jining Medical College, Jining 272100, China
| | - Wen-Jun Zhao
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Huai-Qiang Hu
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Hong-Wei Guo
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Ting Zhang
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Ya-Cheng Lu
- Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China
| | - Shan Jin
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Xiao-Ling Wang
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China
| | - Bing-Zhen Cao
- Department of Neurology, the 960th Hospital of PLA, Jinan 250031, China.
| | - Zhe Wang
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China.
| | - Tan Ding
- Institute of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China; Department of Anatomy, Histology and Embryology, The Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
17
|
Prowse ENP, Chaudhary AR, Sharon D, Hendricks AG. Huntingtin S421 phosphorylation increases kinesin and dynein engagement on early endosomes and lysosomes. Biophys J 2023; 122:1168-1184. [PMID: 36772794 PMCID: PMC10111264 DOI: 10.1016/j.bpj.2023.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/20/2022] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Huntingtin (HTT) is a scaffolding protein that recruits motor proteins to vesicular cargoes, enabling it to regulate kinesin-1, dynein, and myosin-VI-dependent transport. To maintain the native stoichiometry of HTT with its interacting partners, we used CRISPR/Cas9 to induce a phosphomimetic mutation of the endogenous HTT at S421 (HTT-S421D). Using single-particle tracking, optical tweezers, and immunofluorescence, we examined the effects of this mutation on the motility of early endosomes and lysosomes. In HTT-S421D cells, lysosomes exhibit longer displacements and higher processive fractions compared with wild-type (HTT-WT) cells. Kinesins and dyneins exert greater forces on early endosomes and lysosomes in cells expressing HTT-S421D. In addition, endosomes bind to microtubules faster and are more resistant to detachment under load. The recruitment of kinesins and dyneins to microtubules is enhanced in HTT-S421D cells. In contrast, overexpression of HTT had variable effects on the processivity, displacement, and directional bias of both early endosomes and lysosomes. These data indicate that phosphorylation of the endogenous HTT causes early endosomes and lysosomes to move longer distances and more processively by recruiting and activating both kinesin-1 and dynein.
Collapse
Affiliation(s)
- Emily N P Prowse
- Bioengineering Department, McGill University, Montreal, Quebec, Canada
| | | | - David Sharon
- Bioengineering Department, McGill University, Montreal, Quebec, Canada
| | - Adam G Hendricks
- Bioengineering Department, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Chen X, He E, Su C, Zeng Y, Xu J. Huntingtin-associated protein 1-associated intracellular trafficking in neurodegenerative diseases. Front Aging Neurosci 2023; 15:1100395. [PMID: 36824265 PMCID: PMC9941194 DOI: 10.3389/fnagi.2023.1100395] [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: 11/16/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Huntingtin-associated protein 1 (HAP1), the first identified HTT-binding partner, is highly expressed in the central nervous system, and has been found to associated with neurological diseases. Mounting evidence suggests that HAP1 functions as a component of cargo-motor molecules to bind various proteins and participates in intracellular trafficking. It is known that the failure of intracellular transport is a key contributor to the progression of neurodegenerative disorders (NDs) including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), spinal and bulbar muscular atrophy (SBMA) and spinocerebellar ataxia (SCA). The link between HAP1 and various NDs is supported by growing evidence. This review aims to provide a comprehensive overview of the intracellular trafficking function of HAP1 and its involvement in NDs.
Collapse
Affiliation(s)
- Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China,Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Xingxing Chen, ✉
| | - Enhao He
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei, China,Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jiang Xu
- Hubei Key Laboratory of Nerve Injury and Functional Reconstruction, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,Jiang Xu, ✉
| |
Collapse
|
19
|
Roşianu F, Mihaylov SR, Eder N, Martiniuc A, Claxton S, Flynn HR, Jalal S, Domart MC, Collinson L, Skehel M, Snijders AP, Krause M, Tooze SA, Ultanir SK. Loss of NDR1/2 kinases impairs endomembrane trafficking and autophagy leading to neurodegeneration. Life Sci Alliance 2023; 6:6/2/e202201712. [PMID: 36446521 PMCID: PMC9711861 DOI: 10.26508/lsa.202201712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022] Open
Abstract
Autophagy is essential for neuronal development and its deregulation contributes to neurodegenerative diseases. NDR1 and NDR2 are highly conserved kinases, implicated in neuronal development, mitochondrial health and autophagy, but how they affect mammalian brain development in vivo is not known. Using single and double Ndr1/2 knockout mouse models, we show that only dual loss of Ndr1/2 in neurons causes neurodegeneration. This phenotype was present when NDR kinases were deleted both during embryonic development, as well as in adult mice. Proteomic and phosphoproteomic comparisons between Ndr1/2 knockout and control brains revealed novel kinase substrates and indicated that endocytosis is significantly affected in the absence of NDR1/2. We validated the endocytic protein Raph1/Lpd1, as a novel NDR1/2 substrate, and showed that both NDR1/2 and Raph1 are critical for endocytosis and membrane recycling. In NDR1/2 knockout brains, we observed prominent accumulation of transferrin receptor, p62 and ubiquitinated proteins, indicative of a major impairment of protein homeostasis. Furthermore, the levels of LC3-positive autophagosomes were reduced in knockout neurons, implying that reduced autophagy efficiency mediates p62 accumulation and neurotoxicity. Mechanistically, pronounced mislocalisation of the transmembrane autophagy protein ATG9A at the neuronal periphery, impaired axonal ATG9A trafficking and increased ATG9A surface levels further confirm defects in membrane trafficking, and could underlie the impairment in autophagy. We provide novel insight into the roles of NDR1/2 kinases in maintaining neuronal health.
Collapse
Affiliation(s)
- Flavia Roşianu
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Simeon R Mihaylov
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Noreen Eder
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Antonie Martiniuc
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Suzanne Claxton
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| | - Helen R Flynn
- Mass Spectrometry Proteomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Shamsinar Jalal
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Marie-Charlotte Domart
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, UK
| | - Mark Skehel
- Mass Spectrometry Proteomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Ambrosius P Snijders
- Mass Spectrometry Proteomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Matthias Krause
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
20
|
Pero ME, Chowdhury F, Bartolini F. Role of tubulin post-translational modifications in peripheral neuropathy. Exp Neurol 2023; 360:114274. [PMID: 36379274 PMCID: PMC11320756 DOI: 10.1016/j.expneurol.2022.114274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Peripheral neuropathy is a common disorder that results from nerve damage in the periphery. The degeneration of sensory axon terminals leads to changes or loss of sensory functions, often manifesting as debilitating pain, weakness, numbness, tingling, and disability. The pathogenesis of most peripheral neuropathies remains to be fully elucidated. Cumulative evidence from both early and recent studies indicates that tubulin damage may provide a common underlying mechanism of axonal injury in various peripheral neuropathies. In particular, tubulin post-translational modifications have been recently implicated in both toxic and inherited forms of peripheral neuropathy through regulation of axonal transport and mitochondria dynamics. This knowledge forms a new area of investigation with the potential for developing therapeutic strategies to prevent or delay peripheral neuropathy by restoring tubulin homeostasis.
Collapse
Affiliation(s)
- Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University, New York, USA; Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| | - Farihah Chowdhury
- Department of Pathology and Cell Biology, Columbia University, New York, USA
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
21
|
Early TNF-Dependent Regulation of Excitatory and Inhibitory Synapses on Striatal Direct Pathway Medium Spiny Neurons in the YAC128 Mouse Model of Huntington's Disease. J Neurosci 2023; 43:672-680. [PMID: 36517241 PMCID: PMC9888503 DOI: 10.1523/jneurosci.1655-22.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/07/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by a polyglutamine expansion in the huntingtin gene. Neurodegeneration first occurs in the striatum, accompanied by an elevation in inflammatory cytokines. Using the presymptomatic male YAC128 HD model mouse, we examined the synaptic input onto the striatal medium spiny neurons to look for early changes that precede degeneration. We observed an increase in excitatory synaptic strength, as measured by AMPA/NMDA ratios, specifically on direct pathway D1 receptor expressing medium spiny neurons, with no changes on indirect pathway neurons. The changes in excitation were accompanied by a decrease in inhibitory synaptic strength, as measured by the amplitude of miniature inhibitory synaptic currents. The pro-inflammatory cytokine tumor necrosis factor alpha (TNF) was elevated in the striatum of YAC128 at the ages examined. Critically, the changes in excitatory and inhibitory inputs are both dependent on TNF signaling, as blocking TNF signaling genetically or pharmacological normalized synaptic strength. The observed changes in synaptic function are similar to the changes seen in D1 medium spiny neurons treated with high levels of TNF, suggesting that saturating levels of TNF exist in the striatum even at early stages of HD. The increase in glutamatergic synaptic strength and decrease in inhibitory synaptic strength would increase direct pathway neuronal excitability, which may potentiate excitotoxicity during the progress of HD.SIGNIFICANCE STATEMENT The striatum is the first structure to degenerate in Huntington's disease, but the early changes that presage the degeneration are not well defined. Here we identify early synaptic changes in the YAC128 mouse model of Huntington's disease specifically on a subpopulation of striatal neurons. These neurons have stronger excitatory synapses and weaker inhibitory inputs, and thus would increase the susceptibility to excitotoxicity. These changes are dependent on signaling by the pro-inflammatory cytokine TNFα. TNF is elevated even at early presymptomatic stages, and blocking TNF signaling even acutely will reverse the synaptic changes. This suggests early intervention could be important therapeutically.
Collapse
|
22
|
Kim H, Gomez-Pastor R. HSF1 and Its Role in Huntington's Disease Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1410:35-95. [PMID: 36396925 PMCID: PMC12001818 DOI: 10.1007/5584_2022_742] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW Heat shock factor 1 (HSF1) is the master transcriptional regulator of the heat shock response (HSR) in mammalian cells and is a critical element in maintaining protein homeostasis. HSF1 functions at the center of many physiological processes like embryogenesis, metabolism, immune response, aging, cancer, and neurodegeneration. However, the mechanisms that allow HSF1 to control these different biological and pathophysiological processes are not fully understood. This review focuses on Huntington's disease (HD), a neurodegenerative disease characterized by severe protein aggregation of the huntingtin (HTT) protein. The aggregation of HTT, in turn, leads to a halt in the function of HSF1. Understanding the pathways that regulate HSF1 in different contexts like HD may hold the key to understanding the pathomechanisms underlying other proteinopathies. We provide the most current information on HSF1 structure, function, and regulation, emphasizing HD, and discussing its potential as a biological target for therapy. DATA SOURCES We performed PubMed search to find established and recent reports in HSF1, heat shock proteins (Hsp), HD, Hsp inhibitors, HSF1 activators, and HSF1 in aging, inflammation, cancer, brain development, mitochondria, synaptic plasticity, polyglutamine (polyQ) diseases, and HD. STUDY SELECTIONS Research and review articles that described the mechanisms of action of HSF1 were selected based on terms used in PubMed search. RESULTS HSF1 plays a crucial role in the progression of HD and other protein-misfolding related neurodegenerative diseases. Different animal models of HD, as well as postmortem brains of patients with HD, reveal a connection between the levels of HSF1 and HSF1 dysfunction to mutant HTT (mHTT)-induced toxicity and protein aggregation, dysregulation of the ubiquitin-proteasome system (UPS), oxidative stress, mitochondrial dysfunction, and disruption of the structural and functional integrity of synaptic connections, which eventually leads to neuronal loss. These features are shared with other neurodegenerative diseases (NDs). Currently, several inhibitors against negative regulators of HSF1, as well as HSF1 activators, are developed and hold promise to prevent neurodegeneration in HD and other NDs. CONCLUSION Understanding the role of HSF1 during protein aggregation and neurodegeneration in HD may help to develop therapeutic strategies that could be effective across different NDs.
Collapse
Affiliation(s)
- Hyuck Kim
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
23
|
Sadhu A, Badal KK, Zhao Y, Ali AA, Swarnkar S, Tsaprailis G, Crynen GC, Puthanveettil SV. Short-Term and Long-Term Sensitization Differentially Alters the Composition of an Anterograde Transport Complex in Aplysia. eNeuro 2023; 10:ENEURO.0266-22.2022. [PMID: 36549915 PMCID: PMC9829102 DOI: 10.1523/eneuro.0266-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/09/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022] Open
Abstract
Long-term memory formation requires anterograde transport of proteins from the soma of a neuron to its distal synaptic terminals. This allows new synaptic connections to be grown and existing ones remodeled. However, we do not yet know which proteins are transported to synapses in response to activity and temporal regulation. Here, using quantitative mass spectrometry, we have profiled anterograde protein cargos of a learning-regulated molecular motor protein kinesin [Aplysia kinesin heavy chain 1 (ApKHC1)] following short-term sensitization (STS) and long-term sensitization (LTS) in Aplysia californica Our results reveal enrichment of specific proteins associated with ApKHC1 following both STS and LTS, as well as temporal changes within 1 and 3 h of LTS training. A significant number of proteins enriched in the ApKHC1 complex participate in synaptic function, and, while some are ubiquitously enriched across training conditions, a few are enriched in response to specific training. For instance, factors aiding new synapse formation, such as synaptotagmin-1, dynamin-1, and calmodulin, are differentially enriched in anterograde complexes 1 h after LTS but are depleted 3 h after LTS. Proteins including gelsolin-like protein 2 and sec23A/sec24A, which function in actin filament stabilization and vesicle transport, respectively, are enriched in cargos 3 h after LTS. These results establish that the composition of anterograde transport complexes undergo experience-dependent specific changes and illuminate dynamic changes in the communication between soma and synapse during learning.
Collapse
Affiliation(s)
- Abhishek Sadhu
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Kerriann K Badal
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
- Integrated Biology Graduate Program, Florida Atlantic University, Jupiter, Florida 33458
| | - Yibo Zhao
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Adia A Ali
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Supriya Swarnkar
- Department of Neuroscience, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - George Tsaprailis
- Proteomics Core, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | - Gogce C Crynen
- Bioinformatics Core, UF Scripps Biomedical Research, University of Florida, Jupiter, Florida 33458
| | | |
Collapse
|
24
|
Huntingtin and Other Neurodegeneration-Associated Proteins in the Development of Intracellular Pathologies: Potential Target Search for Therapeutic Intervention. Int J Mol Sci 2022; 23:ijms232415533. [PMID: 36555175 PMCID: PMC9779313 DOI: 10.3390/ijms232415533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are currently incurable. Numerous experimental data accumulated over the past fifty years have brought us closer to understanding the molecular and cell mechanisms responsible for their development. However, these data are not enough for a complete understanding of the genesis of these diseases, nor to suggest treatment methods. It turns out that many cellular pathologies developing during neurodegeneration coincide from disease to disease. These observations give hope to finding a common intracellular target(s) and to offering a universal method of treatment. In this review, we attempt to analyze data on similar cellular disorders among neurodegenerative diseases in general, and polyglutamine neurodegenerative diseases in particular, focusing on the interaction of various proteins involved in the development of neurodegenerative diseases with various cellular organelles. The main purposes of this review are: (1) to outline the spectrum of common intracellular pathologies and to answer the question of whether it is possible to find potential universal target(s) for therapeutic intervention; (2) to identify specific intracellular pathologies and to speculate about a possible general approach for their treatment.
Collapse
|
25
|
Chen X, Sun Y, Chen L, Chen XS, Pan M, Zhang Y, Wang Q, Yang W, Yin P, He D, Guo X, Yang S, Zeng Y, Yan S, Li XJ, Li S. Differential expression and roles of Huntingtin and Huntingtin-associated protein 1 in the mouse and primate brains. Cell Mol Life Sci 2022; 79:554. [PMID: 36251080 PMCID: PMC11802989 DOI: 10.1007/s00018-022-04577-8] [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: 08/03/2022] [Revised: 09/12/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Huntingtin-associated protein 1 (HAP1) is the first identified protein whose function is affected by its abnormal interaction with mutant huntingtin (mHTT), which causes Huntington disease. However, the expression patterns of Hap1 and Htt in the rodent brain are not correlated. Here we found that the primate HAP1, unlike the rodent Hap1, is correlatively expressed with HTT in the primate brains. CRISPR/Cas9 targeting revealed that HAP1 deficiency in the developing human neurons did not affect neuronal differentiation and gene expression as seen in the mouse neurons. However, deletion of HAP1 exacerbated neurotoxicity of mutant HTT in the organotypic brain slices of adult monkeys. These findings demonstrate differential HAP1 expression and function in the mouse and primate brains, and suggest that interaction of HAP1 with mutant HTT may be involved in mutant HTT-mediated neurotoxicity in adult primate neurons.
Collapse
Affiliation(s)
- Xingxing Chen
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, 430000, Hubei, China
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yize Sun
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Laiqiang Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiu-Sheng Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Mingtian Pan
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yiran Zhang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Qi Wang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Weili Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Peng Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Dajian He
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiangyu Guo
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Su Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Wuhan University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
26
|
Irfan Z, Khanam S, Karmakar V, Firdous SM, El Khier BSIA, Khan I, Rehman MU, Khan A. Pathogenesis of Huntington's Disease: An Emphasis on Molecular Pathways and Prevention by Natural Remedies. Brain Sci 2022; 12:1389. [PMID: 36291322 PMCID: PMC9599635 DOI: 10.3390/brainsci12101389] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/25/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Huntington's disease is an inherited autosomal dominant trait neuro-degenerative disorder caused by changes (mutations) of a gene called huntingtin (htt) that is located on the short arm (p) of chromosome 4, CAG expansion mutation. It is characterized by unusual movements, cognitive and psychiatric disorders. OBJECTIVE This review was undertaken to apprehend biological pathways of Huntington's disease (HD) pathogenesis and its management by nature-derived products. Natural products can be lucrative for the management of HD as it shows protection against HD in pre-clinical trials. Advanced research is still required to assess the therapeutic effectiveness of the known organic products and their isolated compounds in HD experimental models. SUMMARY Degeneration of neurons in Huntington's disease is distinguished by progressive loss of motor coordination and muscle function. This is due to the expansion of CAG trinucleotide in the first exon of the htt gene responsible for neuronal death and neuronal network degeneration in the brain. It is believed that the factors such as molecular genetics, oxidative stress, excitotoxicity, mitochondrial dysfunction, neuroglia dysfunction, protein aggregation, and altered UPS leads to HD. The defensive effect of the natural product provides therapeutic efficacy against HD. Recent reports on natural drugs have enlightened the protective role against HD via antioxidant, anti-inflammatory, antiapoptotic, and neurofunctional regulation.
Collapse
Affiliation(s)
- Zainab Irfan
- Department of Pharmaceutical Technology, Brainware University, Kolkata 700125, West Bengal, India
| | - Sofia Khanam
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Howrah 711316, West Bengal, India
| | - Varnita Karmakar
- Department of Pharmacology, Eminent College of Pharmaceutical Technology, Barasat 700126, West Bengal, India
| | - Sayeed Mohammed Firdous
- Department of Pharmacology, Calcutta Institute of Pharmaceutical Technology & AHS, Howrah 711316, West Bengal, India
| | | | - Ilyas Khan
- Department of Mathematics, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Muneeb U. Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
27
|
Gicking AM, Ma TC, Feng Q, Jiang R, Badieyan S, Cianfrocco MA, Hancock WO. Kinesin-1, -2, and -3 motors use family-specific mechanochemical strategies to effectively compete with dynein during bidirectional transport. eLife 2022; 11:e82228. [PMID: 36125250 PMCID: PMC9545524 DOI: 10.7554/elife.82228] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/19/2022] [Indexed: 12/03/2022] Open
Abstract
Bidirectional cargo transport in neurons requires competing activity of motors from the kinesin-1, -2, and -3 superfamilies against cytoplasmic dynein-1. Previous studies demonstrated that when kinesin-1 attached to dynein-dynactin-BicD2 (DDB) complex, the tethered motors move slowly with a slight plus-end bias, suggesting kinesin-1 overpowers DDB but DDB generates a substantial hindering load. Compared to kinesin-1, motors from the kinesin-2 and -3 families display a higher sensitivity to load in single-molecule assays and are thus predicted to be overpowered by dynein complexes in cargo transport. To test this prediction, we used a DNA scaffold to pair DDB with members of the kinesin-1, -2, and -3 families to recreate bidirectional transport in vitro, and tracked the motor pairs using two-channel TIRF microscopy. Unexpectedly, we find that when both kinesin and dynein are engaged and stepping on the microtubule, kinesin-1, -2, and -3 motors are able to effectively withstand hindering loads generated by DDB. Stochastic stepping simulations reveal that kinesin-2 and -3 motors compensate for their faster detachment rates under load with faster reattachment kinetics. The similar performance between the three kinesin transport families highlights how motor kinetics play critical roles in balancing forces between kinesin and dynein, and emphasizes the importance of motor regulation by cargo adaptors, regulatory proteins, and the microtubule track for tuning the speed and directionality of cargo transport in cells.
Collapse
Affiliation(s)
- Allison M Gicking
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Tzu-Chen Ma
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Qingzhou Feng
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Rui Jiang
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| | - Somayesadat Badieyan
- Department of Biological Chemistry and the Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - Michael A Cianfrocco
- Department of Biological Chemistry and the Life Sciences Institute, University of Michigan-Ann ArborAnn ArborUnited States
| | - William O Hancock
- Department of Biomedical Engineering, Pennsylvania State UniversityUniversity ParkUnited States
| |
Collapse
|
28
|
Wennagel D, Braz BY, Capizzi M, Barnat M, Humbert S. Huntingtin coordinates dendritic spine morphology and function through cofilin-mediated control of the actin cytoskeleton. Cell Rep 2022; 40:111261. [PMID: 36044862 DOI: 10.1016/j.celrep.2022.111261] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 06/22/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Compelling evidence indicates that in Huntington's disease (HD), mutation of huntingtin (HTT) alters several aspects of early brain development such as synaptogenesis. It is not clear to what extent the partial loss of wild-type HTT function contributes to these abnormalities. Here we investigate the function of HTT in the formation of spines. Although larger spines normally correlate with more synaptic activity, cell-autonomous depletion of HTT leads to enlarged spines but reduced excitatory synaptic function. We find that HTT is required for the proper turnover of endogenous actin and to recruit AMPA receptors at active synapses; loss of HTT leads to LIM kinase (LIMK) hyperactivation, which maintains cofilin in its inactive state. HTT therefore influences actin dynamics through the LIMK-cofilin pathway. Loss of HTT uncouples spine structure from synaptic function, which may contribute to the ultimate development of HD symptoms.
Collapse
Affiliation(s)
- Doris Wennagel
- University Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, Bâtiment Edmond J. Safra, Chemin Fortuné Ferrini, 38000 Grenoble, La Tronche, France
| | - Barbara Yael Braz
- University Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, Bâtiment Edmond J. Safra, Chemin Fortuné Ferrini, 38000 Grenoble, La Tronche, France
| | - Mariacristina Capizzi
- University Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, Bâtiment Edmond J. Safra, Chemin Fortuné Ferrini, 38000 Grenoble, La Tronche, France; Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - Monia Barnat
- University Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, Bâtiment Edmond J. Safra, Chemin Fortuné Ferrini, 38000 Grenoble, La Tronche, France
| | - Sandrine Humbert
- University Grenoble Alpes, Inserm, U1216, Grenoble Institute Neurosciences, Bâtiment Edmond J. Safra, Chemin Fortuné Ferrini, 38000 Grenoble, La Tronche, France; Institut du Cerveau-Paris Brain Institute (ICM), Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
29
|
White A, McGlone A, Gomez-Pastor R. Protein Kinase CK2 and Its Potential Role as a Therapeutic Target in Huntington's Disease. Biomedicines 2022; 10:1979. [PMID: 36009526 PMCID: PMC9406209 DOI: 10.3390/biomedicines10081979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Huntington's Disease (HD) is a devastating neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the HTT gene, for which no disease modifying therapies are currently available. Much of the recent research has focused on developing therapies to directly lower HTT expression, and while promising, these therapies have presented several challenges regarding administration and efficacy. Another promising therapeutic approach is the modulation of HTT post-translational modifications (PTMs) that are dysregulated in disease and have shown to play a key role in HTT toxicity. Among all PTMs, modulation of HTT phosphorylation has been proposed as an attractive therapeutic option due to the possibility of orally administering specific kinase effectors. One of the kinases described to participate in HTT phosphorylation is Protein Kinase CK2. CK2 has recently emerged as a target for the treatment of several neurological and psychiatric disorders, although its role in HD remains controversial. While pharmacological studies in vitro inhibiting CK2 resulted in reduced HTT phosphorylation and increased toxicity, genetic approaches in mouse models of HD have provided beneficial effects. In this review we discuss potential therapeutic approaches related to the manipulation of HTT-PTMs with special emphasis on the role of CK2 as a therapeutic target in HD.
Collapse
Affiliation(s)
| | | | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
30
|
Gloor Y, Matthey A, Sobo K, Mouterde M, Kosek E, Pickering G, Poloni ES, Cedraschi C, Ehret G, Desmeules JA. Uncovering a Genetic Polymorphism Located in Huntingtin Associated Protein 1 in Modulation of Central Pain Sensitization Signaling Pathways. Front Neurosci 2022; 16:807773. [PMID: 35837121 PMCID: PMC9274135 DOI: 10.3389/fnins.2022.807773] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/27/2022] [Indexed: 11/18/2022] Open
Abstract
Fibromyalgia syndrome (FMS) is characterized by widespread pain and increased sensitivity to nociceptive stimulus or tenderness. While familial aggregation could suggest a potential hereditary component in FMS development, isolation of genetic determinants has proven difficult due to the multi-factorial nature and complexity of the syndrome. Central sensitization is thought to be one of the key mechanisms leading to FMS in a subset of patients. Enhanced central pain signaling can be measured using the Nociceptive Flexion Reflex (NFR) or RIII threshold. We performed a genome-wide association study (GWAS) using an array to genotype 258,756 human genetic polymorphisms in 225 FMS patients and 77 healthy volunteers and searched for genetic variants associated with a lowered NFR threshold. We have identified a potential association between a single nucleotide polymorphism resulting in a common non-synonymous coding mutation in the Huntingtin associated protein 1 (HAP1) gene (rs4796604, MAF = 0.5) and the NFR threshold (p = 4.78E−06). The Hap1 protein is involved in trafficking and is particularly enriched in neurons. Our results suggest a possible involvement of the neuronal trafficking protein HAP1 in modulating pain signaling pathways and thus participate in the establishment of the NFR threshold.
Collapse
Affiliation(s)
- Yvonne Gloor
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
- *Correspondence: Yvonne Gloor,
| | - Alain Matthey
- Clinical Investigation Unit, Clinical Research Center (CRC), Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Komla Sobo
- Division of Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Médéric Mouterde
- Anthropology Unit, Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Eva Kosek
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Gisèle Pickering
- Clinical Investigation Center, Inserm 1405, Centre Hospitalier Universitaire, Clermont-Ferrand, France
- Clinical Investigation Center, Inserm 1405, University Hospital, Clermont-Ferrand, France
| | - Estella S. Poloni
- Anthropology Unit, Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Geneva, Switzerland
| | - Christine Cedraschi
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Georg Ehret
- Division of Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Jules A. Desmeules
- Division of Clinical Pharmacology and Toxicology, Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, Geneva University Hospitals (HUG), Geneva, Switzerland
- Clinical Investigation Unit, Clinical Research Center (CRC), Geneva University Hospitals (HUG), Geneva, Switzerland
| |
Collapse
|
31
|
Selective motor activation in organelle transport along axons. Nat Rev Mol Cell Biol 2022; 23:699-714. [DOI: 10.1038/s41580-022-00491-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
|
32
|
Gupta S, Bazargani N, Drew J, Howden JH, Modi S, Al Awabdh S, Marie H, Attwell D, Kittler JT. The non-adrenergic imidazoline-1 receptor protein nischarin is a key regulator of astrocyte glutamate uptake. iScience 2022; 25:104127. [PMID: 35434559 PMCID: PMC9010640 DOI: 10.1016/j.isci.2022.104127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 12/24/2021] [Accepted: 03/17/2022] [Indexed: 12/02/2022] Open
Abstract
Astrocytic GLT-1 is the main glutamate transporter involved in glutamate buffering in the brain, pivotal for glutamate removal at excitatory synapses to terminate neurotransmission and for preventing excitotoxicity. We show here that the surface expression and function of GLT-1 can be rapidly modulated through the interaction of its N-terminus with the nonadrenergic imidazoline-1 receptor protein, Nischarin. The phox domain of Nischarin is critical for interaction and internalization of surface GLT-1. Using live super-resolution imaging, we found that glutamate accelerated Nischarin-GLT-1 internalization into endosomal structures. The surface GLT-1 level increased in Nischarin knockout astrocytes, and this correlated with a significant increase in transporter uptake current. In addition, Nischarin knockout in astrocytes is neuroprotective against glutamate excitotoxicity. These data provide new molecular insights into regulation of GLT-1 surface level and function and suggest new drug targets for the treatment of neurological disorders. Nischarin phox domain interacts with the N-terminus of the glutamate transporter, GLT-1 Nischarin promotes internalization of GLT-1 to endosomes Glutamate modulates GLT-1 surface levels by regulating the Nischarin-GLT-1 interaction Nischarin loss enhances GLT-1 surface levels, transport currents, and neuroprotection
Collapse
Affiliation(s)
- Swati Gupta
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Narges Bazargani
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - James Drew
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Jack H. Howden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Souvik Modi
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Sana Al Awabdh
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Hélène Marie
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
| | - Josef T. Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, WC1E 6BT London, UK
- Corresponding author
| |
Collapse
|
33
|
Hazim RA, Williams DS. Microtubule Motor Transport of Organelles in a Specialized Epithelium: The RPE. Front Cell Dev Biol 2022; 10:852468. [PMID: 35309899 PMCID: PMC8930850 DOI: 10.3389/fcell.2022.852468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
The retinal pigment epithelium (RPE) is a uniquely polarized epithelium that lies adjacent to the photoreceptor cells in the retina, and is essential for photoreceptor function and viability. Two major motile organelles present in the RPE are the melanosomes, which are important for absorbing stray light, and phagosomes that result from the phagocytosis of the distal tips of the photoreceptor cilium, known as the photoreceptor outer segment (POS). These organelles are transported along microtubules, aligned with the apical-basal axis of the RPE. Although they undergo a directional migration, the organelles exhibit bidirectional movements, indicating both kinesin and dynein motor function in their transport. Apical melanosome localization requires dynein; it has been suggested that kinesin contribution might be complex with the involvement of more than one type of kinesin. POS phagosomes undergo bidirectional movements; roles of both plus- and minus-end directed motors appear to be important in the efficient degradation of phagosomes. This function is directly related to retinal health, with defects in motor proteins, or in the association of the phagosomes with the motors, resulting in retinal degenerative pathologies.
Collapse
Affiliation(s)
- Roni A. Hazim
- Department of Ophthalmology and Stein Eye Institute, Los Angeles, CA, United States
| | - David S. Williams
- Department of Ophthalmology and Stein Eye Institute, Los Angeles, CA, United States
- Department of Neurobiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
34
|
Peris L, Parato J, Qu X, Soleilhac JM, Lanté F, Kumar A, Pero ME, Martínez-Hernández J, Corrao C, Falivelli G, Payet F, Gory-Fauré S, Bosc C, Blanca Ramirez M, Sproul A, Brocard J, Di Cara B, Delagrange P, Buisson A, Goldberg Y, Moutin MJ, Bartolini F, Andrieux A. OUP accepted manuscript. Brain 2022; 145:2486-2506. [PMID: 35148384 PMCID: PMC9337816 DOI: 10.1093/brain/awab436] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/04/2021] [Accepted: 10/26/2021] [Indexed: 11/17/2022] Open
Abstract
Microtubules play fundamental roles in the maintenance of neuronal processes and in synaptic function and plasticity. While dynamic microtubules are mainly composed of tyrosinated tubulin, long-lived microtubules contain detyrosinated tubulin, suggesting that the tubulin tyrosination/detyrosination cycle is a key player in the maintenance of microtubule dynamics and neuronal homeostasis, conditions that go awry in neurodegenerative diseases. In the tyrosination/detyrosination cycle, the C-terminal tyrosine of α-tubulin is removed by tubulin carboxypeptidases and re-added by tubulin tyrosine ligase (TTL). Here we show that TTL heterozygous mice exhibit decreased tyrosinated microtubules, reduced dendritic spine density and both synaptic plasticity and memory deficits. We further report decreased TTL expression in sporadic and familial Alzheimer’s disease, and reduced microtubule dynamics in human neurons harbouring the familial APP-V717I mutation. Finally, we show that synapses visited by dynamic microtubules are more resistant to oligomeric amyloid-β peptide toxicity and that expression of TTL, by restoring microtubule entry into spines, suppresses the loss of synapses induced by amyloid-β peptide. Together, our results demonstrate that a balanced tyrosination/detyrosination tubulin cycle is necessary for the maintenance of synaptic plasticity, is protective against amyloid-β peptide-induced synaptic damage and that this balance is lost in Alzheimer’s disease, providing evidence that defective tubulin retyrosination may contribute to circuit dysfunction during neurodegeneration in Alzheimer’s disease.
Collapse
Affiliation(s)
- Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Julie Parato
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Natural Sciences, SUNY ESC, Brooklyn, NY 11201, USA
| | - Xiaoyi Qu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jean Marc Soleilhac
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Fabien Lanté
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Atul Kumar
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Maria Elena Pero
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy
| | - José Martínez-Hernández
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Charlotte Corrao
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Giulia Falivelli
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Floriane Payet
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sylvie Gory-Fauré
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Christophe Bosc
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marian Blanca Ramirez
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Andrew Sproul
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jacques Brocard
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | | | | | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Yves Goldberg
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Marie Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Francesca Bartolini
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Annie Andrieux
- Univ. Grenoble Alpes, Inserm, U1216, CEA, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| |
Collapse
|
35
|
Garcia JD, Gookin SE, Crosby KC, Schwartz SL, Tiemeier E, Kennedy MJ, Dell'Acqua ML, Herson PS, Quillinan N, Smith KR. Stepwise disassembly of GABAergic synapses during pathogenic excitotoxicity. Cell Rep 2021; 37:110142. [PMID: 34936876 PMCID: PMC8824488 DOI: 10.1016/j.celrep.2021.110142] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/17/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
GABAergic synaptic inhibition controls neuronal firing, excitability, and synaptic plasticity to regulate neuronal circuits. Following an acute excitotoxic insult, inhibitory synapses are eliminated, reducing synaptic inhibition, elevating circuit excitability, and contributing to the pathophysiology of brain injuries. However, mechanisms that drive inhibitory synapse disassembly and elimination are undefined. We find that inhibitory synapses are disassembled in a sequential manner following excitotoxicity: GABAARs undergo rapid nanoscale rearrangement and are dispersed from the synapse along with presynaptic active zone components, followed by the gradual removal of the gephyrin scaffold, prior to complete elimination of the presynaptic terminal. GABAAR nanoscale reorganization and synaptic declustering depends on calcineurin signaling, whereas disassembly of gephyrin relies on calpain activation, and blockade of both enzymes preserves inhibitory synapses after excitotoxic insult. Thus, inhibitory synapse disassembly occurs rapidly, with nanoscale precision, in a stepwise manner and most likely represents a critical step in the progression of hyperexcitability following excitotoxicity.
Collapse
Affiliation(s)
- Joshua D Garcia
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Sara E Gookin
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Kevin C Crosby
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Samantha L Schwartz
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Erika Tiemeier
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Paco S Herson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA; Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Department of Anesthesiology, Neuronal Injury Program, University of Colorado School of Medicine, Anschutz Medical Campus, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Katharine R Smith
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
36
|
Sanyal C, Pietsch N, Ramirez Rios S, Peris L, Carrier L, Moutin MJ. The detyrosination/re-tyrosination cycle of tubulin and its role and dysfunction in neurons and cardiomyocytes. Semin Cell Dev Biol 2021; 137:46-62. [PMID: 34924330 DOI: 10.1016/j.semcdb.2021.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
Among the variety of post-translational modifications to which microtubules are subjected, the detyrosination/re-tyrosination cycle is specific to tubulin. It is conserved by evolution and characterized by the enzymatic removal and re-addition of a gene-encoded tyrosine residue at the C-terminus of α-tubulin. Detyrosinated tubulin can be further converted to Δ2-tubulin by the removal of an additional C-terminal glutamate residue. Detyrosinated and Δ2-tubulin are carried by stable microtubules whereas tyrosinated microtubules are present on dynamic polymers. The cycle regulates trafficking of many cargo transporting molecular motors and is linked to the microtubule dynamics via regulation of microtubule interactions with specific cellular effectors such as kinesin-13. Here, we give an historical overview of the general features discovered for the cycle. We highlight the recent progress toward structure and functioning of the enzymes that keep the levels of tyrosinated and detyrosinated tubulin in cells, the long-known tubulin tyrosine ligase and the recently discovered vasohibin-SVBP complexes. We further describe how the cycle controls microtubule functions in healthy neurons and cardiomyocytes and how deregulations of the cycle are involved in dysfunctions of these highly differentiated cells, leading to neurodegeneration and heart failure in humans.
Collapse
Affiliation(s)
- Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sacnicte Ramirez Rios
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
37
|
Fan R, Lai KO. Understanding how kinesin motor proteins regulate postsynaptic function in neuron. FEBS J 2021; 289:2128-2144. [PMID: 34796656 DOI: 10.1111/febs.16285] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/08/2021] [Accepted: 11/17/2021] [Indexed: 01/07/2023]
Abstract
The Kinesin superfamily proteins (KIFs) are major molecular motors that transport diverse set of cargoes along microtubules to both the axon and dendrite of a neuron. Much of our knowledge about kinesin function is obtained from studies on axonal transport. Emerging evidence reveals how specific kinesin motor proteins carry cargoes to dendrites, including proteins, mRNAs and organelles that are crucial for synapse development and plasticity. In this review, we will summarize the major kinesin motors and their associated cargoes that have been characterized to regulate postsynaptic function in neuron. We will also discuss how specific kinesins are selectively involved in the development of excitatory and inhibitory postsynaptic compartments, their regulation by post-translational modifications (PTM), as well as their roles beyond conventional transport carrier.
Collapse
Affiliation(s)
- Ruolin Fan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kwok-On Lai
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
38
|
Kumar V, Singh C, Singh A. Zebrafish an experimental model of Huntington's disease: molecular aspects, therapeutic targets and current challenges. Mol Biol Rep 2021; 48:8181-8194. [PMID: 34665402 DOI: 10.1007/s11033-021-06787-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Huntington disease (HD) is a lethal autosomal dominant neurodegenerative disease whose exact causative mechanism is still unknown. It can transform from one generation to another generation. The CAG triplet expansion on polyglutamine (PolyQ) tract on Huntingtin protein primarily contributes in HD pathogenesis. Apart from this some another molecular mechanisms are also involved in HD pathology such as loss of Brain derived neurotrophic factor in medium spiny neurons, mitochondrial dysfunction, and alterations in synaptic plasticity are briefly discussed in this review. However, several chemicals (3-nitropropionic acid, and Quinolinic acid) and genetic (mHTT-ΔN17-97Q over expression) experimental models are used to explore the exact pathogenic mechanism and finding of new drug targets for the development of novel therapeutic approaches. The zebrafish (Danio rerio) is widely used in in-vivo screening of several central nervous system (CNS) diseases such as HD, Alzheimer's disease (AD), Parkinson's disease (PD), and in memory deficits. Thus, this makes zebrafish as an excellent animal model for the development of new therapeutic strategies against various CNS disorders. We had reviewed several publications utilizing zebrafish and rodents to explore the disease pathology. Studies suggested that zebrafish genes and their human homologues have conserved functions. Zebrafish advantages and their characteristics over the other experimental animals make it an excellent tool for the disease study. This review explains the possible pathogenic mechanism of HD and also discusses about possible treatment therapies, apart from this we also discussed about possible potential therapeutic targets which will helps in designing of novel therapeutic approaches to overcome the disease progression. Diagrammatic depiction shows prevention of HD pathogenesis through attenuation of various biochemical alterations.
Collapse
Affiliation(s)
- Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Charan Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142001, India
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
39
|
Ghosh A, Singh S. Regulation Of Microtubule: Current Concepts And Relevance To Neurodegenerative Diseases. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:656-679. [PMID: 34323203 DOI: 10.2174/1871527320666210728144043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 02/23/2021] [Indexed: 11/22/2022]
Abstract
Neurodevelopmental disorders (NDDs) are abnormalities linked to neuronal structure and irregularities associated with the proliferation of cells, transportation, and differentiation. NDD also involves synaptic circuitry and neural network alterations known as synaptopathies. Microtubules (MTs) and MTs-associated proteins help to maintain neuronal health as well as their development. The microtubular dynamic structure plays a crucial role in the division of cells and forms mitotic spindles, thus take part in initiating stages of differentiation and polarization for various types of cells. The MTs also take part in the cellular death but MT-based cellular degenerations are not yet well excavated. In the last few years, studies have provided the protagonist activity of MTs in neuronal degeneration. In this review, we largely engrossed our discussion on the change of MT cytoskeleton structure, describing their organization, dynamics, transportation, and their failure causing NDDs. At end of this review, we are targeting the therapeutic neuroprotective strategies on clinical priority and also try to discuss the clues for the development of new MT-based therapy as a new pharmacological intervention. This will be a new potential site to block not only neurodegeneration but also promotes the regeneration of neurons.
Collapse
Affiliation(s)
- Anirban Ghosh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga-142001 Punjab, India
| |
Collapse
|
40
|
Barron JC, Hurley EP, Parsons MP. Huntingtin and the Synapse. Front Cell Neurosci 2021; 15:689332. [PMID: 34211373 PMCID: PMC8239291 DOI: 10.3389/fncel.2021.689332] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Huntington disease (HD) is a monogenic disease that results in a combination of motor, psychiatric and cognitive symptoms. HD is caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene, which results in the production of a pathogenic mutant HTT protein (mHTT). Although there is no cure at present for HD, a number of RNA-targeting therapies have recently entered clinical trials which aim to lower mHTT production through the use of antisense oligonucleotides (ASOs) and RNAi. However, many of these treatment strategies are non-selective in that they cannot differentiate between non-pathogenic wild type HTT (wtHTT) and the mHTT variant. As HD patients are already born with decreased levels of wtHTT, these genetic therapies may result in critically low levels of wtHTT. The consequence of wtHTT reduction in the adult brain is currently under debate, and here we argue that wtHTT loss is not well-tolerated at the synaptic level. Synaptic dysfunction is an extremely sensitive measure of subsequent cell death, and is known to precede neurodegeneration in numerous brain diseases including HD. The present review focuses on the prominent role of wtHTT at the synapse and considers the consequences of wtHTT loss on both pre- and postsynaptic function. We discuss how wtHTT is implicated in virtually all major facets of synaptic neurotransmission including anterograde and retrograde transport of proteins to/from terminal buttons and dendrites, neurotransmitter release, endocytic vesicle recycling, and postsynaptic receptor localization and recycling. We conclude that wtHTT presence is essential for proper synaptic function.
Collapse
Affiliation(s)
- Jessica C Barron
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Emily P Hurley
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| |
Collapse
|
41
|
Rodrigues EC, Grawenhoff J, Baumann SJ, Lorenzon N, Maurer SP. Mammalian Neuronal mRNA Transport Complexes: The Few Knowns and the Many Unknowns. Front Integr Neurosci 2021; 15:692948. [PMID: 34211375 PMCID: PMC8239176 DOI: 10.3389/fnint.2021.692948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
Hundreds of messenger RNAs (mRNAs) are transported into neurites to provide templates for the assembly of local protein networks. These networks enable a neuron to configure different cellular domains for specialized functions. According to current evidence, mRNAs are mostly transported in rather small packages of one to three copies, rarely containing different transcripts. This opens up fascinating logistic problems: how are hundreds of different mRNA cargoes sorted into distinct packages and how are they coupled to and released from motor proteins to produce the observed mRNA distributions? Are all mRNAs transported by the same transport machinery, or are there different adaptors or motors for different transcripts or classes of mRNAs? A variety of often indirect evidence exists for the involvement of proteins in mRNA localization, but relatively little is known about the essential activities required for the actual transport process. Here, we summarize the different types of available evidence for interactions that connect mammalian mRNAs to motor proteins to highlight at which point further research is needed to uncover critical missing links. We further argue that a combination of discovery approaches reporting direct interactions, in vitro reconstitution, and fast perturbations in cells is an ideal future strategy to unravel essential interactions and specific functions of proteins in mRNA transport processes.
Collapse
Affiliation(s)
- Elsa C. Rodrigues
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Julia Grawenhoff
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sebastian J. Baumann
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Nicola Lorenzon
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Sebastian P. Maurer
- Center for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| |
Collapse
|
42
|
Grosso Jasutkar H, Yamamoto A. Do Changes in Synaptic Autophagy Underlie the Cognitive Impairments in Huntington's Disease? J Huntingtons Dis 2021; 10:227-238. [PMID: 33780373 PMCID: PMC8293641 DOI: 10.3233/jhd-200466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although Huntington's disease (HD) is classically considered from the perspective of the motor syndrome, the cognitive changes in HD are prominent and often an early manifestation of disease. As such, investigating the underlying pathophysiology of cognitive changes may give insight into important and early neurodegenerative events. In this review, we first discuss evidence from both HD patients and animal models that cognitive changes correlate with early pathological changes at the synapse, an observation that is similarly made in other neurodegenerative conditions that primarily affect cognition. We then describe how autophagy plays a critical role supporting synaptic maintenance in the healthy brain, and how autophagy dysfunction in HD may thereby lead to impaired synaptic maintenance and thus early manifestations of disease.
Collapse
Affiliation(s)
| | - Ai Yamamoto
- Department of Neurology, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
43
|
Sahin GS, Luis Rodriguez-Llamas J, Dillon C, Medina I, Appleyard SM, Gaiarsa JL, Wayman GA. Leptin increases GABAergic synaptogenesis through the Rho guanine exchange factor β-PIX in developing hippocampal neurons. Sci Signal 2021; 14:14/683/eabe4111. [PMID: 34006608 DOI: 10.1126/scisignal.abe4111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Developing hippocampal neurons undergo rapid synaptogenesis in response to neurotrophic signals to form and refine circuit connections. The adipokine leptin is a satiety factor with neurotrophic actions, which potentiates both glutamatergic and GABAergic synaptogenesis in the hippocampus during neonatal development. Brief exposure to leptin enhances GABAA receptor-dependent synaptic currents in hippocampal neurons. Here, using molecular and electrophysiological techniques, we found that leptin increased the surface localization of GABAA receptors and the number of functional GABAergic synapses in hippocampal cultures from male and female rat pups. Leptin increased the interaction between GABAA receptors and the Rho guanine exchange factor β-PIX (a scaffolding protein at GABAergic postsynaptic sites) in a manner dependent on the kinase CaMKK. We also found that the leptin receptor and β-PIX formed a complex, the amount of which transiently increased upon leptin receptor activation. Furthermore, Tyr985 in the leptin receptor and the SH3 domain of β-PIX are crucial for this interaction, which was required for the developmental increase in GABAergic synaptogenesis. Our results suggest a mechanism by which leptin promotes GABAergic synaptogenesis in hippocampal neurons and reveal further complexity in leptin receptor signaling and its interactome.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jose Luis Rodriguez-Llamas
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Crystal Dillon
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Igor Medina
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Suzanne M Appleyard
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jean-Luc Gaiarsa
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Gary A Wayman
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA.
| |
Collapse
|
44
|
Aiken J, Holzbaur ELF. Cytoskeletal regulation guides neuronal trafficking to effectively supply the synapse. Curr Biol 2021; 31:R633-R650. [PMID: 34033795 DOI: 10.1016/j.cub.2021.02.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The development and proper function of the brain requires the formation of highly complex neuronal circuitry. These circuits are shaped from synaptic connections between neurons and must be maintained over a lifetime. The formation and continued maintenance of synapses requires accurate trafficking of presynaptic and postsynaptic components along the axon and dendrite, respectively, necessitating deliberate and specialized delivery strategies to replenish essential synaptic components. Maintenance of synaptic transmission also requires readily accessible energy stores, produced in part by localized mitochondria, that are tightly regulated with activity level. In this review, we focus on recent developments in our understanding of the cytoskeletal environment of axons and dendrites, examining how local regulation of cytoskeletal dynamics and organelle trafficking promotes synapse-specific delivery and plasticity. These new insights shed light on the complex and coordinated role that cytoskeletal elements play in establishing and maintaining neuronal circuitry.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
45
|
El-Ansary A, Zayed N, Al-Ayadhi L, Qasem H, Anwar M, Meguid NA, Bhat RS, Doşa MD, Chirumbolo S, Bjørklund G. GABA synaptopathy promotes the elevation of caspases 3 and 9 as pro-apoptotic markers in Egyptian patients with autism spectrum disorder. Acta Neurol Belg 2021; 121:489-501. [PMID: 31673995 DOI: 10.1007/s13760-019-01226-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorder (ASD) is classified as a neurodevelopmental disorder characterized by reduced social communication as well as repetitive behaviors. Many studies have proved that defective synapses in ASD influence how neurons in the brain connect and communicate with each other. Synaptopathies arise from alterations that affecting the integrity and/or functionality of synapses and can contribute to synaptic pathologies. This study investigated the GABA levels in plasma being an inhibitory neurotransmitter, caspase 3 and 9 as pro-apoptotic proteins in 20 ASD children and 20 neurotypical controls using the ELISA technique. Analysis of receiver-operating characteristic (ROC) of the data that was obtained to evaluate the diagnostic value of the aforementioned evaluated biomarkers. Pearson's correlations and multiple regressions between the measured variables were also done. While GABA level was reduced in ASD patients, levels of caspases 3 and 9 were significantly higher when compared to neurotypical control participants. ROC and predictiveness curves showed that caspases 3, caspases 9, and GABA might be utilized as predictive markers in autism diagnosis. The present study indicates that the presence of GABAergic dysfunction promotes apoptosis in Egyptian ASD children. The obtained GABA synaptopathies and their connection with apoptosis can both relate to neuronal excitation, and imbalance of the inhibition system, which can be used as reliable predictive biomarkers for ASD.
Collapse
|
46
|
SNX27-Mediated Recycling of Neuroligin-2 Regulates Inhibitory Signaling. Cell Rep 2020; 29:2599-2607.e6. [PMID: 31775031 PMCID: PMC6899438 DOI: 10.1016/j.celrep.2019.10.096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 08/22/2019] [Accepted: 10/24/2019] [Indexed: 01/11/2023] Open
Abstract
GABAA receptors mediate fast inhibitory transmission in the brain, and their number can be rapidly up- or downregulated to alter synaptic strength. Neuroligin-2 plays a critical role in the stabilization of synaptic GABAA receptors and the development and maintenance of inhibitory synapses. To date, little is known about how the amount of neuroligin-2 at the synapse is regulated and whether neuroligin-2 trafficking affects inhibitory signaling. Here, we show that neuroligin-2, when internalized to endosomes, co-localizes with SNX27, a brain-enriched cargo-adaptor protein that facilitates membrane protein recycling. Direct interaction between the PDZ domain of SNX27 and PDZ-binding motif in neuroligin-2 enables membrane retrieval of neuroligin-2, thus enhancing synaptic neuroligin-2 clusters. Furthermore, SNX27 knockdown has the opposite effect. SNX27-mediated up- and downregulation of neuroligin-2 surface levels affects inhibitory synapse composition and signaling strength. Taken together, we show a role for SNX27-mediated recycling of neuroligin-2 in maintenance and signaling of the GABAergic synapse.
Collapse
|
47
|
Gong YJ, Feng Y, Cao YY, Zhao J, Wu W, Zheng YY, Wu JR, Li X, Yang GZ, Zhou X. Huntingtin-associated protein 1 plays an essential role in the pathogenesis of type 2 diabetes by regulating the translocation of GLUT4 in mouse adipocytes. BMJ Open Diabetes Res Care 2020; 8:8/1/e001199. [PMID: 33060070 PMCID: PMC7566288 DOI: 10.1136/bmjdrc-2020-001199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/30/2020] [Accepted: 09/05/2020] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Glucose disposal by insulin-responsive tissues maintains the body glucose homeostasis and insulin resistance leads to a risk of developing type 2 diabetes (T2DM). Insulin stimulates the translocation of glucose transporter isoform 4 (GLUT4) vesicles from intracellular compartments to the plasma membrane to facilitate glucose uptake. However, the underlying mechanisms of GLUT4 vesicle translocation are not well defined. Here we show the role of huntingtin-associated protein 1 (HAP1) in GLUT4 translocation in adipocytes and the pathogenesis of T2DM. RESEARCH DESIGN AND METHODS The parameters for glucose metabolism including body weight, glucose tolerance and insulin tolerance were assessed in wild-type (WT) and Hap1+/- mice. HAP1 protein expression was verified in adipose tissue. Hap1 mRNA and protein expression was monitored in adipose tissue of high-fat diet (HFD)-induced diabetic mice. Insulin-stimulated GLUT4 vesicle translocation and glucose uptake were detected using immunofluorescence techniques and quantified in primary adipocytes from Hap1-/- mice. The interaction between HAP1 and GLUT4 was assessed by immunofluorescence colocalization and co-immunoprecipitation in HEK293 cells and adipose tissue. The role of sortilin in HAP1 and GLUT4 interaction was approved by co-immunoprecipitation and RNA interference. RESULTS The expression of Hap1 mRNA and protein was detected in WT mouse adipose tissue and downregulated in adipose tissue of HFD-induced diabetic mice. Hap1+/- mice exhibited increased body weight, pronounced glucose tolerance and significant insulin intolerance compared with the WT mice. HAP1 colocalized with GLUT4 in mouse adipocytes and cotransfected HEK293 cells. Furthermore, the insulin-stimulated GLUT4 vesicle translocation and glucose uptake were defective in Hap1-/- adipocytes. Finally, sortilin mediated the interaction of HAP1 and GLUT4. CONCLUSIONS Our study showed that HAP1 formed a protein complex with GLUT4 and sortilin, and played a critical role in insulin-stimulated GLUT4 translocation in adipocytes. Its downregulation may contribute to the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Yan-Ju Gong
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yuan-Yuan Cao
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jia Zhao
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wu
- Institute of Biology, National Institute of Measurement and Testing Technology, Chengdu, Sichuan, China
| | - Ya-Yun Zheng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jia-Rui Wu
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xin Li
- Department of Pathophysiology, School of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Gui-Zhi Yang
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xue Zhou
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Svrzikapa N, Longo KA, Prasad N, Boyanapalli R, Brown JM, Dorset D, Yourstone S, Powers J, Levy SE, Morris AJ, Vargeese C, Goyal J. Investigational Assay for Haplotype Phasing of the Huntingtin Gene. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:162-173. [PMID: 33209959 PMCID: PMC7648085 DOI: 10.1016/j.omtm.2020.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/04/2020] [Indexed: 01/20/2023]
Abstract
Novel treatments for Huntington's disease (HD), a progressive neurodegenerative disorder, include selective targeting of the mutant allele of the huntingtin gene (mHTT) carrying the abnormally expanded disease-causing cytosine-adenine-guanine (CAG) repeat. WVE-120101 and WVE-120102 are investigational stereopure antisense oligonucleotides that enable selective suppression of mHTT by targeting single-nucleotide polymorphisms (SNPs) that are in haplotype phase with the CAG repeat expansion. Recently developed long-read sequencing technologies can capture CAG expansions and distant SNPs of interest and potentially facilitate haplotype-based identification of patients for clinical trials of oligonucleotide therapies. However, improved methods are needed to phase SNPs with CAG repeat expansions directly and reliably without need for familial genotype/haplotype data. Our haplotype phasing method uses single-molecule real-time sequencing and a custom algorithm to determine with confidence bases at SNPs on mutant alleles, even without familial data. Herein, we summarize this methodology and validate the approach using patient-derived samples with known phasing results. Comparison of experimentally measured CAG repeat lengths, heterozygosity, and phasing with previously determined results showed improved performance. Our methodology enables the haplotype phasing of SNPs of interest and the disease-causing, expanded CAG repeat of the huntingtin gene, enabling accurate identification of patients with HD eligible for allele-selective clinical studies.
Collapse
Affiliation(s)
- Nenad Svrzikapa
- Wave Life Sciences Ltd., Cambridge, MA 02138, USA.,Department of Paediatrics, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | | | - Nripesh Prasad
- HudsonAlpha Discovery, Discovery Life Sciences, Huntsville, AL 35806, USA.,Genomic Services Laboratory, HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | | | - Daniel Dorset
- HudsonAlpha Discovery, Discovery Life Sciences, Huntsville, AL 35806, USA.,Genomic Services Laboratory, HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Jason Powers
- Q Solutions
- EA Genomics, LLC, Morrisville, NC 27560, USA
| | - Shawn E Levy
- HudsonAlpha Discovery, Discovery Life Sciences, Huntsville, AL 35806, USA.,Genomic Services Laboratory, HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | | | - Jaya Goyal
- Wave Life Sciences Ltd., Cambridge, MA 02138, USA
| |
Collapse
|
49
|
Lopes AT, Hausrat TJ, Heisler FF, Gromova KV, Lombino FL, Fischer T, Ruschkies L, Breiden P, Thies E, Hermans-Borgmeyer I, Schweizer M, Schwarz JR, Lohr C, Kneussel M. Spastin depletion increases tubulin polyglutamylation and impairs kinesin-mediated neuronal transport, leading to working and associative memory deficits. PLoS Biol 2020; 18:e3000820. [PMID: 32866173 PMCID: PMC7485986 DOI: 10.1371/journal.pbio.3000820] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/11/2020] [Accepted: 08/10/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations in the gene encoding the microtubule-severing protein spastin (spastic paraplegia 4 [SPG4]) cause hereditary spastic paraplegia (HSP), associated with neurodegeneration, spasticity, and motor impairment. Complicated forms (complicated HSP [cHSP]) further include cognitive deficits and dementia; however, the etiology and dysfunctional mechanisms of cHSP have remained unknown. Here, we report specific working and associative memory deficits upon spastin depletion in mice. Loss of spastin-mediated severing leads to reduced synapse numbers, accompanied by lower miniature excitatory postsynaptic current (mEPSC) frequencies. At the subcellular level, mutant neurons are characterized by longer microtubules with increased tubulin polyglutamylation levels. Notably, these conditions reduce kinesin-microtubule binding, impair the processivity of kinesin family protein (KIF) 5, and reduce the delivery of presynaptic vesicles and postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Rescue experiments confirm the specificity of these results by showing that wild-type spastin, but not the severing-deficient and disease-associated K388R mutant, normalizes the effects at the synaptic, microtubule, and transport levels. In addition, short hairpin RNA (shRNA)-mediated reduction of tubulin polyglutamylation on spastin knockout background normalizes KIF5 transport deficits and attenuates the loss of excitatory synapses. Our data provide a mechanism that connects spastin dysfunction with the regulation of kinesin-mediated cargo transport, synapse integrity, and cognition. This study identifies deficits in working and associative memory in spastin knockout mice, resembling the cognitive deficits described in humans with severe forms of SPG4-type hereditary spastic paraplegia. Mechanistically, the findings suggest that impaired microtubule growth, kinesin motility and cargo delivery of synaptic AMPA receptors may contribute to the etiology of the disease.
Collapse
Affiliation(s)
- André T. Lopes
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torben J. Hausrat
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frank F. Heisler
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kira V. Gromova
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Franco L. Lombino
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Fischer
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Laura Ruschkies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Petra Breiden
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Edda Thies
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Transgenic Animal Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology Unit, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jürgen R. Schwarz
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Division of Neurophysiology, University of Hamburg, Hamburg, Germany
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
50
|
Proteotoxicity and Neurodegenerative Diseases. Int J Mol Sci 2020; 21:ijms21165646. [PMID: 32781742 PMCID: PMC7460676 DOI: 10.3390/ijms21165646] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/01/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases are a major burden for our society, affecting millions of people worldwide. A main goal of past and current research is to enhance our understanding of the mechanisms underlying proteotoxicity, a common theme among these incurable and debilitating conditions. Cell proteome alteration is considered to be one of the main driving forces that triggers neurodegeneration, and unraveling the biological complexity behind the affected molecular pathways constitutes a daunting challenge. This review summarizes the current state on key processes that lead to cellular proteotoxicity in Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, providing a comprehensive landscape of recent literature. A foundational understanding of how proteotoxicity affects disease etiology and progression may provide essential insight towards potential targets amenable of therapeutic intervention.
Collapse
|