1
|
Yadav DK, Ahoulou EO, Anderson DE, Bej A, Hell JW, Ames JB. L-Type Voltage-Gated Ca 2+ Channel C-Terminal Proximal and Distal Domains (PCRD and DCRD) Bind to the IQ-Motif and May Modulate Channel Function. Biochemistry 2025; 64:1933-1942. [PMID: 40238689 DOI: 10.1021/acs.biochem.4c00880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
The L-type voltage-gated Ca2+ channel (CaV1.2) controls gene expression, cardiac contraction, and neuronal activity. The C-terminal cytosolic region of the CaV1.2 alpha subunit (α1C) contains two domains known as proximal and distal C-terminal regulatory domains (PCRD and DCRD), which have been suggested to control Ca2+-dependent channel inactivation (CDI). Previous studies identified a salt bridge interaction between PCRD and DCRD that might be central to channel function. In this study, we expressed and purified recombinant constructs of PCRD (residues 1680-1750) and DCRD (residues 2035-2089) in E. coli for NMR structural analysis. PCRD and DCRD each exhibit backbone NMR chemical shifts consistent with a random coil and lack of tertiary structure. A disordered random coil structure may explain the absence of electron density for these domains in recent cryo-EM structures of CaV1.2. Despite this lack of structure, fluorescence polarization binding assays reveal PCRD and DCRD each bind to the CaV1.2 IQ-motif (residues 1644-1668) with dissociation constants of 1.5 ± 1 and 14 ± 5 μM, respectively. Both PCRD and DCRD also become insoluble in the presence of high micromolar levels of the IQ peptide, consistent with each domain forming an insoluble complex with the IQ peptide. AlphaFold3 predicts that DCRD adopts a 3-helix bundle that binds to the helical IQ-motif, while PCRD was previously suggested to form a 4-helix bundle. We propose that the PCRD and DCRD bind to opposite sides of the helical IQ-motif, which may oppose calmodulin (CaM) binding to CaV1.2 and thus modulate channel CDI.
Collapse
Affiliation(s)
- Deepak Kumar Yadav
- Department of Chemistry, University of California, Davis, California95616, United States
| | - Effibe O Ahoulou
- Department of Chemistry, University of California, Davis, California95616, United States
| | - David E Anderson
- Department of Chemistry, University of California, Davis, California95616, United States
| | - Aritra Bej
- Department of Chemistry, University of California, Davis, California95616, United States
- Department of Pharmacology, University of California, Davis, California95616, United States
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, California95616, United States
| | - James B Ames
- Department of Chemistry, University of California, Davis, California95616, United States
| |
Collapse
|
2
|
Chen J, Dong X, Lin Y, Lv C. The critical role of GLP-1 signaling pathways in the pathology of Parkinson's disease and diabetes. Pathol Res Pract 2025; 270:155985. [PMID: 40279852 DOI: 10.1016/j.prp.2025.155985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/18/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
This review assesses the roles of GLP-1 and its receptor agonists (GLP-1RAs) in the treatment of diabetes and Parkinson's disease, integrating current theories and research. GLP-1, a vital endogenous hormone, regulates insulin secretion, delays gastric emptying, and promotes satiety, showing significant potential for diabetes management. However, its brief lifespan and restricted blood-brain barrier penetration limit its clinical application. To overcome these constraints, researchers have developed GLP-1 receptor agonists that prolong its action and exhibit high efficacy in diabetes treatment. Recent studies further reveal GLP-1's neuroprotective effects, notably its potential in managing neurodegenerative disorders such as Parkinson's disease. GLP-1RAs mitigate neuroinflammation, reduce oxidative stress, and enhance neuroprotection, suggesting substantial potential for treating neurodegenerative diseases. Additionally, to enhance GLP-1RAs' efficacy in the nervous system, researchers have introduced novel drug delivery approaches, including nanoparticle carriers and molecular modifications, to improve stability and targeting accuracy. In conclusion, this review comprehensively analyzes the mechanisms, clinical applications, and challenges of GLP-1 and its receptor agonists in managing diabetes and Parkinson's disease, while identifying future research and clinical opportunities.
Collapse
Affiliation(s)
- Jinhao Chen
- China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Xiang Dong
- China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | | | - Cunming Lv
- China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, Hubei 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
3
|
Sánchez-Cárdenas C, Oliver EI, Chávez JC, Luque GM, Hernández-Cruz A, Buffone MG, Darszon A, Visconti PE, Romarowski A. Ion channels and transporters involved in calcium flux regulation in mammalian sperm. Curr Top Dev Biol 2025; 162:351-385. [PMID: 40180515 DOI: 10.1016/bs.ctdb.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
After ejaculation, mammalian spermatozoa are not capable of fertilizing a metaphase II-arrested egg. They require to undergo a series of biochemical and physiological processes collectively known as capacitation. In all these processes, the regulation of calcium ions fluxes plays essential roles and involves participation of many channels and transporters localized in the plasma membrane as well as in the membrane of intracellular organelles. In mammalian sperm, a fraction of these molecules has been proposed to contribute to mature sperm function. However, in many cases, the evidence for the presence of a given protein is based on the use of agonists and antagonists with more than one target. In this review, we will critically analyze the published evidence supporting the presence of these molecules in mammalian sperm with special emphasis to methods involving tandem mass spectrometry identification, electrophysiological evidence and controlled immunoassays.
Collapse
Affiliation(s)
- Claudia Sánchez-Cárdenas
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico.
| | - Enrique I Oliver
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Julio C Chávez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Guillermina M Luque
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Arturo Hernández-Cruz
- Departamento de Neuropatología Molecular y Laboratorio Nacional de Canalopatías, Instituto de Fisiología Celular UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, UNAM, Cuernavaca, Mexico
| | - Pablo E Visconti
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States.
| | - Ana Romarowski
- Instituto de Biología y Medicina Experimental (IBYME), National Research Council of Argentina (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
4
|
Limpitikul WB, Dick IE. Inactivation of CaV1 and CaV2 channels. J Gen Physiol 2025; 157:e202313531. [PMID: 39883005 PMCID: PMC11781272 DOI: 10.1085/jgp.202313531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/02/2025] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) are highly expressed throughout numerous biological systems and play critical roles in synaptic transmission, cardiac excitation, and muscle contraction. To perform these various functions, VGCCs are highly regulated. Inactivation comprises a critical mechanism controlling the entry of Ca2+ through these channels and constitutes an important means to regulate cellular excitability, shape action potentials, control intracellular Ca2+ levels, and contribute to long-term potentiation and depression. For CaV1 and CaV2 channel families, inactivation proceeds via two distinct processes. Voltage-dependent inactivation (VDI) reduces Ca2+ entry through the channel in response to sustained or repetitive depolarization, while Ca2+-dependent inactivation (CDI) occurs in response to elevations in intracellular Ca2+ levels. These processes are critical for physiological function and undergo exquisite fine-tuning through multiple mechanisms. Here, we review known determinants and modulatory features of these two critical forms of channel regulation and their role in normal physiology and pathophysiology.
Collapse
Affiliation(s)
| | - Ivy E. Dick
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
5
|
Romanovsky E, Choudhary A, Peles D, Abu-Akel A, Stern S. Uncovering convergence and divergence between autism and schizophrenia using genomic tools and patients' neurons. Mol Psychiatry 2025; 30:1019-1028. [PMID: 39237719 PMCID: PMC11835745 DOI: 10.1038/s41380-024-02740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/07/2024]
Abstract
Autism spectrum disorders (ASDs) are highly heritable and result in abnormal repetitive behaviors and impairment in communication and cognitive skills. Previous studies have focused on the genetic correlation between ASDs and other neuropsychiatric disorders, but an in-depth understanding of the correlation to other disorders is required. We conducted an extensive meta-analysis of common variants identified in ASDs by genome-wide association studies (GWAS) and compared it to the consensus genes and single nucleotide polymorphisms (SNPs) of Schizophrenia (SCZ). We found approximately 75% of the GWAS genes that are associated with ASD are also associated with SCZ. We further investigated the cellular phenotypes of neurons derived from induced pluripotent stem cell (iPSC) models in ASD and SCZ. Our findings revealed that ASD and SCZ neurons initially follow divergent developmental trajectories compared to control neurons. However, despite these early diametrical differences, both ASD and SCZ neurons ultimately display similar deficits in synaptic activity as they mature. This significant genetic overlap between ASD and SCZ, coupled with the convergence towards similar synaptic deficits, highlights the intricate interplay of genetic and developmental factors in shaping the shared underlying mechanisms of these complex neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Eva Romanovsky
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - David Peles
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ahmad Abu-Akel
- School of Psychological Sciences, University of Haifa, Haifa, Israel
- The Haifa Brain and Behavior Hub, University of Haifa, Haifa, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel.
| |
Collapse
|
6
|
Bogacheva PO, Potapova DA, Gaydukov AE. Sortilin and L-type Calcium Channels May be Involved in the Unusual Mechanism of proBDNF Signaling in Regenerating Mouse Neuromuscular Junctions. Neurochem Res 2025; 50:104. [PMID: 39998597 DOI: 10.1007/s11064-025-04360-8] [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: 10/17/2024] [Revised: 01/17/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
proBDNF and its main proteolytic product BDNF play crucial roles in maturation of neuromuscular junctions during development or reinnervation. We investigated the mechanisms of acute proBDNF effects on synaptic transmission in mouse motor synapses regenerating after nerve crush. The cleavage-resistant proBDNF mimicked the previously shown effect of cleavable proBDNF- GIRK-mediated decrease in the miniature endplate potential (MEPP) frequency accompanied by slight hyperpolarization of postsynaptic membrane. Remarkably, this effect did not utilize canonical proBDNF signaling pathway since inhibition of either p75 receptors with LM11A-31 or sortilin with AF38469 was not able to prevent it. Without sortilin activity, proBDNF downregulated the quantal content of multiquantal endplate potentials (EPP). This non-canonical action of proneurotrophin via TrkB receptors highlights the important role of sortilin as a safeguard preventing the spread of the negative effect of proBDNF on the evoked neurotransmitter release in regenerating motor synapses. In the absence of sortilin activity L-type calcium channels emerged as the key players providing proBDNF-induced decrease of EPP quantal content, while they were not involved in proBDNF-induced decrease of MEPP frequency. Sortilin-independent but TrkB- and GIRK-mediated inhibition of spontaneous release by proBDNF was not associated with the activity of acetylcholine (M2) or purinergic (A1 and P2Y13) metabotropic receptors. We propose that depending on sortilin involvement, proBDNF selectively affects spontaneous or evoked quantal neurotransmitter release via different branches of signaling pathway that ensure the presynaptic activation of GIRK or L-type calcium channels, respectively.
Collapse
Affiliation(s)
- P O Bogacheva
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation.
| | - D A Potapova
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - A E Gaydukov
- Faculty of Biology, Department of Human and Animal Physiology, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
7
|
Zhang L, Chu Q, Jiang S, Shao B. Genetic evidence for amlodipine's protective role in gastroesophageal reflux disease: A focus on CACNB2. PLoS One 2025; 20:e0309805. [PMID: 39965006 PMCID: PMC11835245 DOI: 10.1371/journal.pone.0309805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/20/2024] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE This study aims to elucidate the causal relationship between genetically predicted amlodipine use and the risk of gastroesophageal reflux disease (GERD) using a bidirectional Mendelian Randomization (MR) approach and to explore the underlying genetic and molecular mechanisms through functional enrichment analysis and the construction of a competing endogenous RNA (ceRNA) network. METHODS Publicly available GWAS datasets from the Neale Lab consortium were used, including data on amlodipine (13,693 cases, 323,466 controls) and GERD (14,316 cases, 322,843 controls). Genome-wide significant SNPs were selected as instrumental variables and clustered by linkage disequilibrium. MR analysis was conducted using R software with all five methods. Sensitivity analyses assessed pleiotropy and heterogeneity. Drug target genes were analyzed using GO and KEGG pathways. GeneMANIA was used for network visualization, and a ceRNA network was constructed with Cytoscape. Differential gene expression analysis on GERD-related datasets from GEO validated the findings. RESULTS The MR analysis indicated a significant negative association between genetically predicted amlodipine use and GERD risk (IVW OR = 0.872, 95% CI = 0.812-0.937, P = 0.0002). Sensitivity analyses confirmed the robustness of these findings, showing no evidence of pleiotropy or heterogeneity. The enrichment analysis identified key biological processes and pathways involving calcium ion transport and signaling. The ceRNA network highlighted core targets such as CACNB2, which were further validated by differential expression analysis intersecting drug target genes with GERD-related gene expression changes. CONCLUSION This study provides robust evidence of a protective effect of amlodipine against GERD, supported by genetic and molecular analyses. The findings suggest that calcium channel blockers like amlodipine could be repurposed for GERD treatment. The identification of CACNB2 and other core targets in the ceRNA network offers novel insights into the pathophysiology of GERD and potential therapeutic targets, paving the way for personalized medicine approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Liuzhao Zhang
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Quanwang Chu
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Shuyue Jiang
- Department of Critical Care Medicine, Anhui Jing’an Medicine Hospital, Hefei, China
| | - Bo Shao
- Department of Pathology, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
8
|
Zhao Y, Cai Y, Wang W, Bai Y, Liu M, Wang Y, Niu W, Luo Z, Xia L, Zhu J, Zhao F, Tay FR, Niu L. Periosteum-bone inspired hierarchical scaffold with endogenous piezoelectricity for neuro-vascularized bone regeneration. Bioact Mater 2025; 44:339-353. [PMID: 39512423 PMCID: PMC11541236 DOI: 10.1016/j.bioactmat.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 11/15/2024] Open
Abstract
The development of scaffolds for repairing critical-sized bone defects heavily relies on establishing a neuro-vascularized network for proper penetration of nerves and blood vessels. Despite significant advancements in using artificial bone-like scaffolds infused with various agents, challenges remain. Natural bone tissue consists of a porous bone matrix surrounded by a neuro-vascularized periosteum, with unique piezoelectric properties essential for bone growth. Drawing inspiration from this assembly, we developed a periosteum-bone-mimicking bilayer scaffold with piezoelectric properties for regeneration of critical-sized bone defects. The periosteum-like layer of this scaffold features a double network hydrogel composed of chelated alginate, gelatin methacrylate, and sintered whitlockite nanoparticles, emulating the viscoelastic and piezoelectric properties of the natural periosteum. The bone-like layer is composed of a porous structure of chitosan and bioactive hydroxyapatite created through a biomineralization process. Unlike conventional bone-like scaffolds, this bioinspired bilayer scaffold significantly enhances osteogenesis, angiogenesis, and neurogenesis combined with low-intensity pulsed ultrasound-assisted piezoelectric stimulation. Such a scheme enhances neuro-vascularized bone regeneration in vivo. The results suggest that the bilayer scaffold could serve as an effective self-powered electrical stimulator to expedite bone regeneration under dynamic physical stimulation.
Collapse
Affiliation(s)
- Yao Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yunfan Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, PR China
| | - Wenkai Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, PR China
| | - Yongkang Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Mingyi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Yan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Wen Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Zhixiao Luo
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, PR China
| | - Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, PR China
| | - Juanfang Zhu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, PR China
| | - Fei Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | - Franklin R. Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Lina Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| |
Collapse
|
9
|
Guo SJ, Shi YQ, Zheng YN, Liu H, Zheng YL. The Voltage-Gated Calcium Channel α2δ Subunit in Neuropathic Pain. Mol Neurobiol 2025; 62:2561-2572. [PMID: 39136907 DOI: 10.1007/s12035-024-04424-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/06/2024] [Indexed: 01/28/2025]
Abstract
Neuropathic pain (NP) is a chronic pain caused by injury or disease of the somatosensory nervous system, or it can be directly caused by disease. It often presents with clinical features like spontaneous pain, hyperalgesia, and dysesthesia. At present, voltage-gated calcium ion channels (VGCCs) are known to be closely related to the development of NP, especially the α2δ subunit. The α2δ subunit is a regulatory subunit of VGCCs. It exists mainly in the brain and peripheral nervous system, especially in nerve cells, and it plays a crucial part in regulating presynaptic and postsynaptic functions. Furthermore, the α2δ subunit influences neuronal excitation and pain signaling by promoting its expression and localization through binding to VGCC-related subunits. The α2δ subunit is widely used in the management of NP as a target of antiepileptic drugs gabapentin and pregabalin. Although drug therapy is one of the treatments for NP, its clinical application is limited due to the adverse reactions caused by drug therapy. Therefore, further research on the therapeutic target α2δ subunit is needed, and attempts are made to obtain an effective treatment for relieving NP without side effects. This review describes the current associated knowledge on the function of the α2δ subunit in perceiving and modulating NP.
Collapse
Affiliation(s)
- Sheng-Jie Guo
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Yu-Qin Shi
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Ya-Nan Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Hui Liu
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China
| | - Yi-Li Zheng
- Department of Sport Rehabilitation, Shanghai University of Sport, 399 Changhai Road, Yangpu District, Shanghai, China.
- Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China.
| |
Collapse
|
10
|
Ren Z, Li T, Liu X, Zhang Z, Chen X, Chen W, Li K, Sheng J. Transforming growth factor-beta 1 enhances discharge activity of cortical neurons. Neural Regen Res 2025; 20:548-556. [PMID: 38819066 PMCID: PMC11317929 DOI: 10.4103/nrr.nrr-d-23-00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/12/2023] [Accepted: 11/22/2023] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00031/figure1/v/2024-05-28T214302Z/r/image-tiff Transforming growth factor-beta 1 (TGF-β1) has been extensively studied for its pleiotropic effects on central nervous system diseases. The neuroprotective or neurotoxic effects of TGF-β1 in specific brain areas may depend on the pathological process and cell types involved. Voltage-gated sodium channels (VGSCs) are essential ion channels for the generation of action potentials in neurons, and are involved in various neuroexcitation-related diseases. However, the effects of TGF-β1 on the functional properties of VGSCs and firing properties in cortical neurons remain unclear. In this study, we investigated the effects of TGF-β1 on VGSC function and firing properties in primary cortical neurons from mice. We found that TGF-β1 increased VGSC current density in a dose- and time-dependent manner, which was attributable to the upregulation of Nav1.3 expression. Increased VGSC current density and Nav1.3 expression were significantly abolished by preincubation with inhibitors of mitogen-activated protein kinase kinase (PD98059), p38 mitogen-activated protein kinase (SB203580), and Jun NH2-terminal kinase 1/2 inhibitor (SP600125). Interestingly, TGF-β1 significantly increased the firing threshold of action potentials but did not change their firing rate in cortical neurons. These findings suggest that TGF-β1 can increase Nav1.3 expression through activation of the ERK1/2-JNK-MAPK pathway, which leads to a decrease in the firing threshold of action potentials in cortical neurons under pathological conditions. Thus, this contributes to the occurrence and progression of neuroexcitatory-related diseases of the central nervous system.
Collapse
Affiliation(s)
- Zhihui Ren
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Tian Li
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Xueer Liu
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Zelin Zhang
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Xiaoxuan Chen
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Weiqiang Chen
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| | - Jiangtao Sheng
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong Province, China
| |
Collapse
|
11
|
López-Carrillo J, Bernáldez-Sarabia J, Pawar TJ, Jiménez S, Dueñas S, Figueroa-Montiel A, Olivares-Romero JL, Granados-Soto V, Licea-Navarro AF, Caram-Salas NL. Systemic antihyperalgesic effect of a novel conotoxin from Californiconus californicus in an inflammatory pain model. FRONTIERS IN PAIN RESEARCH 2025; 5:1500789. [PMID: 39925365 PMCID: PMC11802583 DOI: 10.3389/fpain.2024.1500789] [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: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025] Open
Abstract
Introduction This study explores the analgesic potential of the novel conotoxin O1_cal6.4b, derived from Californiconus californicus, as a candidate for pain management in a model of inflammatory pain. Methods O1_cal6.4b was systemically administered to Wistar rats, and its effects on thermal hyperalgesia and motor coordination were evaluated. Comparative analyses were conducted against O1_cal6.4d, ω-MVIIA, and standard analgesics (morphine, dexamethasone, and diclofenac). Structural differences between O1_cal6.4b and O1_cal6.4d were examined using in silico modeling and molecular dynamics simulations. Results Systemic administration of O1_cal6.4b significantly reduced thermal hyperalgesia in a dose-dependent manner without impairing motor coordination. The analgesic effect of O1_cal6.4b was superior to that of O1_cal6.4d, ω-MVIIA, and standard analgesics. Structural analyses revealed notable differences between O1_cal6.4b and O1_cal6.4d, suggesting unique functional properties. Discussion The findings indicate that O1_cal6.4b exhibits a promising analgesic profile with advantages over traditional opioid-based therapies. These results underscore the molecular diversity of conotoxins and highlight their potential as innovative analgesic treatments. Further research is needed to elucidate the mechanism of action of this novel conotoxin.
Collapse
Affiliation(s)
| | | | - Tushar J. Pawar
- Red de Estudios Moleculares Avanzados, Instituto de Ecología (INECOL), Xalapa, Mexico
| | - Samanta Jiménez
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | - Salvador Dueñas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
| | | | | | - Vinicio Granados-Soto
- Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, South Campus, Mexico City, Mexico
| | - Alexei F. Licea-Navarro
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- Dirección de Impulso a la Innovación y el Desarrollo (DIID), CICESE, Ensenada, Mexico
| | - Nadia L. Caram-Salas
- Departamento de Innovación Biomédica, CICESE, Ensenada, Mexico
- CONAHCYT. Av. Insurgentes Sur 1582, Col. Crédito Constructor, Deleg Benito Juárez, Mexico City, Mexico
| |
Collapse
|
12
|
Morris VS, Richards EMB, Morris R, Dart C, Helassa N. Structure-Function Diversity of Calcium-Binding Proteins (CaBPs): Key Roles in Cell Signalling and Disease. Cells 2025; 14:152. [PMID: 39936944 PMCID: PMC11816674 DOI: 10.3390/cells14030152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Calcium (Ca2+) signalling is a fundamental cellular process, essential for a wide range of physiological functions. It is regulated by various mechanisms, including a diverse family of Ca2+-binding proteins (CaBPs), which are structurally and functionally similar to calmodulin (CaM). The CaBP family consists of six members (CaBP1, CaBP2, CaBP4, CaBP5, CaBP7, and CaBP8), each exhibiting unique localisation, structural features, and functional roles. In this review, we provide a structure-function analysis of the CaBP family, highlighting the key similarities and differences both within the family and in comparison to CaM. It has been shown that CaBP1-5 share similar structural and interaction characteristics, while CaBP7 and CaBP8 form a distinct subfamily with unique properties. This review of current CaBP knowledge highlights the critical gaps in our understanding, as some CaBP members are less well characterised than others. We also examine pathogenic mutations within CaBPs and their functional impact, showing the need for further research to improve treatment options for associated disorders.
Collapse
Affiliation(s)
| | | | | | | | - Nordine Helassa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3BX, UK; (V.S.M.); (E.M.B.R.); (R.M.); (C.D.)
| |
Collapse
|
13
|
Chen H, Fang HQ, Liu JT, Chang SY, Cheng LB, Sun MX, Feng JR, Liu ZM, Zhang YH, Rosen CJ, Liu P. Atlas of Fshr expression from novel reporter mice. eLife 2025; 13:RP93413. [PMID: 39773308 PMCID: PMC11709436 DOI: 10.7554/elife.93413] [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] [Indexed: 01/11/2025] Open
Abstract
The FSH-FSHR pathway has been considered an essential regulator in reproductive development and fertility. But there has been emerging evidence of FSHR expression in extragonadal organs. This poses new questions and long-term debates regarding the physiological role of the FSH-FSHR, and underscores the need for reliable, in vivo analysis of FSHR expression in animal models. However, conventional methods have proven insufficient for examining FSHR expression due to several limitations. To address this challenge, we developed Fshr-ZsGreen reporter mice under the control of Fshr endogenous promoter using CRISPR-Cas9. With this novel genetic tool, we provide a reliable readout of Fshr expression at single-cell resolution level in vivo and in real time. Reporter animals were also subjected to additional analyses,to define the accurate expression profile of FSHR in gonadal and extragonadal organs/tissues. Our compelling results not only demonstrated Fshr expression in intragonadal tissues but also, strikingly, unveiled notably increased expression in Leydig cells, osteoblast lineage cells, endothelial cells in vascular structures, and epithelial cells in bronchi of the lung and renal tubes. The genetic decoding of the widespread pattern of Fshr expression highlights its physiological relevance beyond reproduction and fertility, and opens new avenues for therapeutic options for age-related disorders of the bones, lungs, kidneys, and hearts, among other tissues. Exploiting the power of the Fshr knockin reporter animals, this report provides the first comprehensive genetic record of the spatial distribution of FSHR expression, correcting a long-term misconception about Fshr expression and offering prospects for extensive exploration of FSH-FSHR biology.
Collapse
Affiliation(s)
- Hongqian Chen
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Hui-Qing Fang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Department of Dentistry, The 980th Hospital of the PLA Joint Logistic Support ForceShijiazhuangChina
| | - Jin-Tao Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Shi-Yu Chang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Li-Ben Cheng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ming-Xin Sun
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Jian-Rui Feng
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| | - Ze-Min Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | - Yong-Hong Zhang
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
- Shanxi Medical Universityersity, The Second Hospital, University Shanxi Medical UniversityTaiyuanChina
| | | | - Peng Liu
- Laboratory of Bone and Adipose Biology, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
14
|
Qi Z, Chen X, Zhu Y, Yue Q, Ji W. Electrochemical sensing of transient ascorbate fluctuation under hypoxic stress in live rat brain. Talanta 2025; 282:126996. [PMID: 39383720 DOI: 10.1016/j.talanta.2024.126996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
Hypoxia, a common cause of programmed cell death or apoptosis, represents a neuropathological process. Although certain response proteins to hypoxic stress and their effects on cell status and fate have been identified, the real-time quantification of smaller neurochemicals to understand pathogenic mechanism in live rat brain during such stress remains unexplored. In this study, by employing a cutting-edge electrochemical tool developed with carbon nanotube-sheathed carbon fiber microelectrode that offers remarkable selectivity and temporal/spatial resolution for monitoring ascorbate, we observed a substantial efflux of ascorbate in response to hypoxic stress in live rat brain. Furthermore, using a small molecule compound as channel inhibitor to investigate the behavior of ascorbate efflux, we found that this efflux is closely correlated with N-methyl-D-aspartic acid receptor-induced neuronal excitability. Notably, antagonistic actions on volume-sensitive anion channel can suppress ascorbate efflux evoked by hypoxic stress, further revealing that ascorbate fluctuation is volume-sensitive anion channel-dependent. This research not only facilitates a greater understanding of the neurochemical mechanism in hypoxia but also uncovers a potential biomarker for future closed-loop therapies.
Collapse
Affiliation(s)
- Ziyang Qi
- School of Education and Psychology, University of Jinan, Jinan, 250022, China
| | - Xingshuai Chen
- School of Physical Education, University of Jinan, Jinan, 250022, China
| | - Ye Zhu
- Shenzhen Research Institute of Shandong University, Shenzhen, 518000, China
| | - Qingwei Yue
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China.
| | - Wenliang Ji
- School of Physical Education, University of Jinan, Jinan, 250022, China.
| |
Collapse
|
15
|
Tian S, Ma C, Zhu Y, Xu Q, Wu J, Qiu Y, Liang T, Ren G, Huang Z, Sun X, Kong L, Wei X, Yu Z, Wang P, Wan H. A light-addressable potentiometric sensor-based extracellular calcium dynamic monitoring and imaging platform for cellular calcium channel drug evaluation. Biosens Bioelectron 2025; 267:116814. [PMID: 39362138 DOI: 10.1016/j.bios.2024.116814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
Disruption and dysregulation of cellular calcium channel function can lead to diseases such as ischemic stroke, heart failure, and arrhythmias. Corresponding calcium channel drugs typically require preliminary efficacy evaluations using in vitro models such as cells and simulated tissues before clinical testing. However, traditional detection and evaluation methods often encounter challenges in long-term continuous monitoring and lack calcium specificity. In this study, a dynamic monitoring system based on ion-sensitive membranes for light-addressable potentiometric sensor (LAPS) was developed to meet the demand for monitoring changes in extracellular calcium ion (Ca2+) concentration in live cells. The effects of Ca2+ channel agonists and blockers on 2D and 3D HL-1 cells were investigated, with changes in extracellular Ca2+ concentration reflecting cellular calcium metabolism, facilitating drug evaluation. Additionally, calcium imaging technology with optical addressing capability complemented the LAPS system's ability to perceive 3D cell morphology, enhancing its drug evaluation capabilities. This work provides a novel, label-free, specific, and stable technique for monitoring cellular calcium metabolism. It achieves both continuous monitoring at single points and custom sensing area calcium imaging, holding significant implications for drug screening and disease treatment related to human calcium homeostasis.
Collapse
Affiliation(s)
- Shichao Tian
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chiyu Ma
- Xi'an Institute of Applied Optics, Xi'an, 710065, China.
| | - Yuxuan Zhu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qihui Xu
- National Key Laboratory of Wide Bandgap Semiconductor Devices and Integrated Technology, Xidian University, 710071, China
| | - Jianguo Wu
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Yong Qiu
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| | - Tao Liang
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Guangqing Ren
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhuoru Huang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xianyou Sun
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liubing Kong
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xinwei Wei
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhengyin Yu
- State Key Laboratory of Transducer Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Hao Wan
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, China; Binjiang Institute of Zhejiang University, Hangzhou, 310053, China.
| |
Collapse
|
16
|
Balbinot G, Milosevic M, Morshead CM, Iwasa SN, Zariffa J, Milosevic L, Valiante TA, Hoffer JA, Popovic MR. The mechanisms of electrical neuromodulation. J Physiol 2025; 603:247-284. [PMID: 39740777 DOI: 10.1113/jp286205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 11/20/2024] [Indexed: 01/02/2025] Open
Abstract
The central and peripheral nervous systems are specialized to conduct electrical currents that underlie behaviour. When this multidimensional electrical system is disrupted by degeneration, damage, or disuse, externally applied electrical currents may act to modulate neural structures and provide therapeutic benefit. The administration of electrical stimulation can exert precise and multi-faceted effects at cellular, circuit and systems levels to restore or enhance the functionality of the central nervous system by providing an access route to target specific cells, fibres of passage, neurotransmitter systems, and/or afferent/efferent communication to enable positive changes in behaviour. Here we examine the neural mechanisms that are thought to underlie the therapeutic effects seen with current neuromodulation technologies. To gain further insights into the mechanisms associated with electrical stimulation, we summarize recent findings from genetic dissection studies conducted in animal models. KEY POINTS: Electricity is everywhere around us and is essential for how our nerves communicate within our bodies. When nerves are damaged or not working properly, using exogenous electricity can help improve their function at distinct levels - inside individual cells, within neural circuits, and across entire systems. This method can be tailored to target specific types of cells, nerve fibres, neurotransmitters and communication pathways, offering significant therapeutic potential. This overview explains how exogenous electricity affects nerve function and its potential benefits, based on research in animal studies. Understanding these effects is important because electrical neuromodulation plays a key role in medical treatments for neurological conditions.
Collapse
Affiliation(s)
- Gustavo Balbinot
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
| | - Matija Milosevic
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Cindi M Morshead
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
- KITE Research Institute - University Health Network, Toronto, ON, Canada
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Stephanie N Iwasa
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
- KITE Research Institute - University Health Network, Toronto, ON, Canada
| | - Jose Zariffa
- KITE Research Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
- Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - Luka Milosevic
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
- KITE Research Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Taufik A Valiante
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
- KITE Research Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - Joaquín Andrés Hoffer
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Milos R Popovic
- Center for Advancing Neurotechnological Innovation to Application - CRANIA, University Health Network, Toronto, ON, Canada
- KITE Research Institute - University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
- Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
17
|
Zheng Y, Ju Y, Liu Y, Yang F. Piezoelectric Nanoparticle-Based Ultrasound Wireless Piezoelectric Neuromodulation Inhibits Epileptiform Activity of Primary Neurons. ACS APPLIED BIO MATERIALS 2024; 7:8543-8551. [PMID: 39556669 DOI: 10.1021/acsabm.4c01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Piezoelectric materials, renowned for their ability to convert mechanical energy into electrical energy, have gained attention for their potential in biomedical applications. In particular, piezoelectric nanoparticles, such as barium titanate nanoparticles, hold great promise for treating neurologically related diseases. In this study, barium titanate piezoelectric nanoparticles are used as stimulators to directly treat epileptic neurons. After being modified by polyethylene glycol, barium titanate nanoparticles have shown excellent biocompatibility and dispersibility. Furthermore, such nanoparticles offer wireless piezoelectric stimulation to neurons in response to low-intensity pulsed ultrasound. More importantly, our experiments reveal that piezoelectric stimulation immediately reduces neuronal intracellular calcium concentration and restores cell viability. These effects are attributed to the opening of voltage-gated calcium channels and the release of active substances. These findings offer insights into the potential of piezoelectric stimulation as an approach for epilepsy treatment and enhance our understanding of the mechanisms underlying electrical stimulation in epileptic neurons.
Collapse
Affiliation(s)
- Yuxiang Zheng
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Yongxu Ju
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Yang Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, PR China
| |
Collapse
|
18
|
Pikor D, Hurła M, Słowikowski B, Szymanowicz O, Poszwa J, Banaszek N, Drelichowska A, Jagodziński PP, Kozubski W, Dorszewska J. Calcium Ions in the Physiology and Pathology of the Central Nervous System. Int J Mol Sci 2024; 25:13133. [PMID: 39684844 DOI: 10.3390/ijms252313133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Calcium ions play a key role in the physiological processes of the central nervous system. The intracellular calcium signal, in nerve cells, is part of the neurotransmission mechanism. They are responsible for stabilizing membrane potential and controlling the excitability of neurons. Calcium ions are a universal second messenger that participates in depolarizing signal transduction and contributes to synaptic activity. These ions take an active part in the mechanisms related to memory and learning. As a result of depolarization of the plasma membrane or stimulation of receptors, there is an extracellular influx of calcium ions into the cytosol or mobilization of these cations inside the cell, which increases the concentration of these ions in neurons. The influx of calcium ions into neurons occurs via plasma membrane receptors and voltage-dependent ion channels. Calcium channels play a key role in the functioning of the nervous system, regulating, among others, neuronal depolarization and neurotransmitter release. Channelopathies are groups of diseases resulting from mutations in genes encoding ion channel subunits, observed including the pathophysiology of neurological diseases such as migraine. A disturbed ability of neurons to maintain an appropriate level of calcium ions is also observed in such neurodegenerative processes as Alzheimer's disease, Parkinson's disease, Huntington's disease, and epilepsy. This review focuses on the involvement of calcium ions in physiological and pathological processes of the central nervous system. We also consider the use of calcium ions as a target for pharmacotherapy in the future.
Collapse
Affiliation(s)
- Damian Pikor
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Mikołaj Hurła
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Bartosz Słowikowski
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Oliwia Szymanowicz
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Joanna Poszwa
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Natalia Banaszek
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Alicja Drelichowska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Paweł P Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Wojciech Kozubski
- Chair and Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
| |
Collapse
|
19
|
Sun Y, Geng J, Fan Y, Li Y, Zhong Y, Cai J, Liu X, Wang S, Gong Y, Chang C, Yang Y, Fan C. A Non-Invasive and DNA-free Approach to Upregulate Mammalian Voltage-Gated Calcium Channels and Neuronal Calcium Signaling via Terahertz Stimulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405436. [PMID: 39435751 DOI: 10.1002/advs.202405436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/22/2024] [Indexed: 10/23/2024]
Abstract
Mammalian voltage-gated calcium channels (CaV) play critical roles in cardiac excitability, synaptic transmission, and gene transcription. Dysfunctions in CaV are implicated in a variety of cardiac and neurodevelopmental disorders. Current pharmacological approaches to enhance CaV activity are limited by off-target effects, drug metabolism issues, cytotoxicity, and imprecise modulation. Additionally, genetically-encoded channel activators and optogenetic tools are restricted by gene delivery challenges and biosafety concerns. Here a novel terahertz (THz) wave-based method to upregulate CaV1.2, a key subtype of CaV, and boost CaV1-mediated Ca2+ signaling in neurons without introducing exogenous DNA is presented. Using molecular dynamics simulations, it is shown that 42.5 THz (7.05 µm, 1418 cm-1) waves enhance Ca2+ conductance in CaV1.2 by resonating with the stretching mode of the -COO- group in the selectivity filter. Electrophysiological recordings and Ca2+ imaging confirm that these waves rapidly, reversibly, and non-thermally increase calcium influx of CaV1.2 in HEK293 cells and induce acute Ca2+ signals in neurons. Furthermore, this irradiation upregulates critical CaV1 signals, including CREB phosphorylation and c-Fos expression, in vitro and in vivo, without raising significant biosafety risks. This DNA-free, non-invasive approach offers a promising approach for modulating CaV gating and Ca2+ signaling and treating diseases characterized by deficits in CaV functions.
Collapse
Affiliation(s)
- Yuankun Sun
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Jinli Geng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Yu Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Yangmei Li
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
| | - Yuan Zhong
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
| | - Jing Cai
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Xiaodong Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Shaomeng Wang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Yubin Gong
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100072, P. R. China
- School of Physics, Peking University, Beijing, 100871, P. R. China
| | - Yaxiong Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University Beijing, Beijing, 100191, P. R. China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
20
|
Haddad S, Hessenberger M, Ablinger C, Eibl C, Stanika R, Campiglio M, Obermair GJ. Autism-Linked Mutations in α 2δ-1 and α 2δ-3 Reduce Protein Membrane Expression but Affect Neither Calcium Channels nor Trans-Synaptic Signaling. Pharmaceuticals (Basel) 2024; 17:1608. [PMID: 39770450 PMCID: PMC11677996 DOI: 10.3390/ph17121608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND α2δ proteins regulate membrane trafficking and biophysical properties of voltage-gated calcium channels. Moreover, they modulate axonal wiring, synapse formation, and trans-synaptic signaling. Several rare missense variants in CACNA2D1 (coding for α2δ-1) and CACNA2D3 (coding for α2δ-3) genes were identified in patients with autism spectrum disorder (ASD). However, the pathogenicity of these variants is not known, and the molecular mechanism by which α2δ proteins may contribute to the pathophysiology of autism is, as of today, not understood. Therefore, in this study we functionally characterized two heterozygous missense variants in α2δ-1 (p.R351T) and α2δ-3 (p.A275T), previously identified in patients with ASD. METHODS Electrophysiological recordings in transfected tsA201 cells were used to study specific channel-dependent functions of mutated α2δ proteins. Membrane expression, presynaptic targeting, and trans-synaptic signaling of mutated α2δ proteins were studied upon expression in murine cultured hippocampal neurons. RESULTS Homologous expression of both mutated α2δ proteins revealed a strongly reduced membrane expression and synaptic localization compared to the corresponding wild type α2δ proteins. Moreover, the A275T mutation in α2δ-3 resulted in an altered glycosylation pattern upon heterologous expression. However, neither of the mutations compromised the biophysical properties of postsynaptic L-type (CaV1.2 and CaV1.3) and presynaptic P/Q-type (CaV2.1) channels when co-expressed in tsA201 cells. Furthermore, presynaptic expression of p.R351T in the α2δ-1 splice variant lacking exon 23 did not affect trans-synaptic signaling to postsynaptic GABAA receptors. CONCLUSIONS Our data provide evidence that the pathophysiological mechanisms of ASD-causing mutations of α2δ proteins may not involve their classical channel-dependent and trans-synaptic functions. Alternatively, these mutations may induce subtle changes in synapse formation or neuronal network function, highlighting the need for future α2δ protein-linked disease models.
Collapse
Affiliation(s)
- Sabrin Haddad
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Manuel Hessenberger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Cornelia Ablinger
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Clarissa Eibl
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Ruslan Stanika
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| | - Marta Campiglio
- Institute of Physiology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Gerald J. Obermair
- Division of Physiology, Department of Pharmacology, Physiology, and Microbiology, Karl Landsteiner University of Health Sciences, 3500 Krems, Austria; (S.H.)
| |
Collapse
|
21
|
Montalbetti N, Manrique-Maldonado G, Ikeda Y, Dalghi M, Kanai A, Apodaca G, Carattino MD. Expression of Acid-Sensing Ion Channel 3 in Afferents Averts Long-Term Sensitization and the Development of Visceral Pain. Int J Mol Sci 2024; 25:12503. [PMID: 39684215 DOI: 10.3390/ijms252312503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/07/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Sensitization of primary afferents is essential for the development of pain, but the molecular events involved in this process and its reversal are poorly defined. Recent studies revealed that acid-sensing ion channels (ASICs) control the excitability of nociceptors in the urinary bladder. Using genetic and pharmacological tools we show that ASICs are functionally coupled with voltage-gated Ca2+ channels to mediate Ca2+ transients evoked by acidification in sensory neurons. Genetic deletion of Asic3 of these sensory neurons does not alter the mechanical response of bladder afferents to distension in naïve mice. Both control and sensory neuron conditional Asic3 knockout (Asic3-KO) mice with chemical cystitis induced by cyclophosphamide (CYP) administration exhibit frequent low volume voiding events. However, these changes are transient and revert over time. Of major significance, in Asic3-KO mice, CYP treatment results in the sensitization of a subset of bladder afferents and pelvic allodynia that persist beyond the resolution of the inflammatory process. Thus, ASICs function is necessary to prevent long-term sensitization of visceral nociceptors.
Collapse
Affiliation(s)
- Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
| | | | - Youko Ikeda
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburg, PA 15261, USA
| | - Marianela Dalghi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
| | - Anthony Kanai
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburg, PA 15261, USA
| | - Gerard Apodaca
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburg, PA 15261, USA
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburg, PA 15261, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburg, PA 15261, USA
| |
Collapse
|
22
|
Wan D, Lu T, Li C, Hu C. Glucocorticoids Rapidly Modulate Ca V1.2-Mediated Calcium Signals through Kv2.1 Channel Clusters in Hippocampal Neurons. J Neurosci 2024; 44:e0179242024. [PMID: 39299804 PMCID: PMC11551909 DOI: 10.1523/jneurosci.0179-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/15/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
The precise regulation of Ca2+ signals plays a crucial role in the physiological functions of neurons. Here, we investigated the rapid effect of glucocorticoids on Ca2+ signals in cultured hippocampal neurons from both female and male rats. In cultured hippocampal neurons, glucocorticoids inhibited the spontaneous somatic Ca2+ spikes generated by Kv2.1-organized Ca2+ microdomains. Furthermore, glucocorticoids rapidly reduced the cell surface expressions of Kv2.1 and CaV1.2 channels in hippocampal neurons. In HEK293 cells transfected with Kv2.1 alone, glucocorticoids significantly reduced the surface expression of Kv2.1 with little effect on K+ currents. In HEK293 cells transfected with CaV1.2 alone, glucocorticoids inhibited CaV1.2 currents but had no effect on the cell surface expression of CaV1.2. Notably, in the presence of wild-type Kv2.1, glucocorticoids caused a decrease in the surface expression of CaV1.2 channels in HEK293 cells. However, this effect was not observed in the presence of nonclustering Kv2.1S586A mutant channels. Live-cell imaging showed that glucocorticoids rapidly decreased Kv2.1 clusters on the plasma membrane. Correspondingly, Western blot results indicated a significant increase in the cytoplasmic level of Kv2.1, suggesting the endocytosis of Kv2.1 clusters. Glucocorticoids rapidly decreased the intracellular cAMP concentration and the phosphorylation level of PKA in hippocampal neurons. The PKA inhibitor H89 mimicked the effect of glucocorticoids on Kv2.1, while the PKA agonist forskolin abrogated the effect. In conclusion, glucocorticoids rapidly suppress CaV1.2-mediated Ca2+ signals in hippocampal neurons by promoting the endocytosis of Kv2.1 channel clusters through reducing PKA activity.
Collapse
Affiliation(s)
- Di Wan
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Tongchuang Lu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Chenyang Li
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| | - Changlong Hu
- Department of Physiology and Neurobiology, School of Life Sciences, Fudan University, Shanghai 200438, P.R.China,
- International Human Phenome Institute (Shanghai), Shanghai 200433, P.R.China
| |
Collapse
|
23
|
Ma XF, Liu AJ, Zheng Z, Hu BX, Zhi YX, Liu C, Tian SJ. Resolving and functional analysis of RNA editing sites in sheep ovaries and associations with litter size. Animal 2024; 18:101342. [PMID: 39471744 DOI: 10.1016/j.animal.2024.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 11/01/2024] Open
Abstract
Sheep litter size is a critical trait in mutton production. While litter size regulation in relation to DNA transcription have been rigorously investigated, the function of RNA editing remains less explored. To elucidate the mechanisms controlling sheep fecundity at the RNA editing level and identify pivotal RNA editing sites, this study scrutinised RNA editing sites (RESs) in follicular and luteal phases of ovaries from sheep with high and low fecundity, and the functions of population-specific RESs were subsequently analysed. A total of 2 182 475 RESs, 74.61% of which were A-to-I and C-to-U sites, were identified. These RESs were fairly evenly dispersed over the chromosomes, with 46.8% showing close clustering (inter-site distance < 300 bp). Notably, 93% were primarily situated in intronic and intergenic regions. In the follicular phase, pivotal RESs were found in the introns of genes including LPS responsive beige-like anchor, MCC regulator of Wnt signalling, and RWD domain containing 3, among others, and in the exon region of EvC ciliary complex subunit 2. In the luteal phase, RESs were observed in the introns of genes such as H/ACA ribonucleoprotein assembly factor and SDA1 domain-containing 1, and the exon and 3'UTR regions of polypeptide N-acetylgalactosaminyltransferase 15 and ilvB acetolactate synthase-like, respectively. High-fecundity sheep showed RESs in the follicular phase in genes such as fibrillin 1, cyclin-dependent kinase 6, and roundabout 1, and in genes such as autophagy-related 2B and versican in the luteal phase. Thirteen RESs specific to the follicular phase and eight specific to the luteal phase were identified in high-fecundity sheep ovaries. These RESs offer promising molecular targets and enhance understanding of multiple births in sheep from the perspective of posttranscriptional alterations.
Collapse
Affiliation(s)
- X F Ma
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - A J Liu
- Department of Agricultural and Animal Husbandry Engineering, Cangzhou Technical College, Hebei, Cangzhou, China
| | - Z Zheng
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - B X Hu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - Y X Zhi
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - C Liu
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China
| | - S J Tian
- College of Animal Science and Technology, Hebei Agricultural University, Hebei, Baoding 071000, China; The Research Center of Cattle and Sheep Embryonic Technique of Hebei Province, Hebei, Baoding, 071000 Baoding, China.
| |
Collapse
|
24
|
Kasper S, Eckert A. Silexan in anxiety, depression, and related disorders: pharmacological background and clinical data. Eur Arch Psychiatry Clin Neurosci 2024:10.1007/s00406-024-01923-8. [PMID: 39453446 DOI: 10.1007/s00406-024-01923-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/31/2024] [Indexed: 10/26/2024]
Abstract
We present a narrative review of clinical trials investigating the anxiolytic and antidepressant effects of silexan, an active substance derived from lavender oil and summarize nonclinical findings from pharmacological studies supporting its therapeutic use. Six studies investigated the efficacy of the lavender oil in patients with subthreshold and generalized anxiety disorders as well as in mixed anxiety and depressive disorder (MADD). Furthermore, we present data indicating that silexan may influence sleep quality as well as anxiety or depressive disorders in individuals with post-COVID-19. Silexan taken orally at a daily dose of 80 mg for 10 weeks was significantly superior to placebo in reducing psychic and somatic symptoms of anxiety and was as effective as 0.5 mg/d lorazepam and 20 mg/d paroxetine. In patients with mild or moderate major depression, silexan was superior to placebo and comparably effective to 50 mg/d sertraline. Significant antidepressant effects were also observed in MADD and depression co-morbid with anxiety. The herbal product had a beneficial effect on activities of daily living and health-related quality of life. Adverse events associated with silexan in clinical trials were limited to eructation and mild, transient gastrointestinal complaints. The herbal product was not associated with drug interactions, sedation, sleep disturbance, dependence and abuse potential, sexual dysfunction, weight gain or withdrawal symptoms. Silexan was therefore safe and effective in subthreshold and syndromal anxiety disorders and in major depression.
Collapse
Affiliation(s)
- Siegfried Kasper
- Department of Molecular Neuroscience, Center of Brain Research, Medical University of Vienna, Spitalgasse 4, Vienna, A-1090, Austria.
| | - Anne Eckert
- Neurobiology Laboratory for Brain Aging and Mental Health, Psychiatric University Clinics Basel, Basel, Switzerland
- Research Platform Molecular and Cognitive Neurosciences, University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
Shabani K, Krupp J, Lemesre E, Lévy N, Tran H. Voltage-Gated Ion Channel Compensatory Effect in DEE: Implications for Future Therapies. Cells 2024; 13:1763. [PMID: 39513870 PMCID: PMC11544952 DOI: 10.3390/cells13211763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Developmental and Epileptic Encephalopathies (DEEs) represent a clinically and genetically heterogeneous group of rare and severe epilepsies. DEEs commonly begin early in infancy with frequent seizures of various types associated with intellectual disability and leading to a neurodevelopmental delay or regression. Disease-causing genomic variants have been identified in numerous genes and are implicated in over 100 types of DEEs. In this context, genes encoding voltage-gated ion channels (VGCs) play a significant role, and part of the large phenotypic variability observed in DEE patients carrying VGC mutations could be explained by the presence of genetic modifier alleles that can compensate for these mutations. This review will focus on the current knowledge of the compensatory effect of DEE-associated voltage-gated ion channels and their therapeutic implications in DEE. We will enter into detailed considerations regarding the sodium channels SCN1A, SCN2A, and SCN8A; the potassium channels KCNA1, KCNQ2, and KCNT1; and the calcium channels CACNA1A and CACNA1G.
Collapse
Affiliation(s)
- Khadijeh Shabani
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| | | | | | | | - Helene Tran
- Institut de Recherches Servier, Rue Francis Perrin, 91190 Gif-sur-Yvette, France; (J.K.); (E.L.); (N.L.)
| |
Collapse
|
26
|
Zhou Q, Li H, Zhang Y, Zhao Y, Wang C, Liu C. Hydrogen-Rich Water to Enhance Exercise Performance: A Review of Effects and Mechanisms. Metabolites 2024; 14:537. [PMID: 39452918 PMCID: PMC11509640 DOI: 10.3390/metabo14100537] [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: 08/27/2024] [Revised: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Hydrogen-rich water (HRW) has garnered significant interest within the sports and exercise science community due to its selective antioxidant properties. Despite its potential benefits, comprehensive reviews specifically addressing its effects on athletic performance are limited. This review aims to assess the impact of HRW on sports performance and explore the underlying molecular biological mechanisms, with the goal of elucidating how HRW might enhance athletic performance. Methods: This review synthesizes research on HRW by examining articles published between 1980 and April 2024 in databases such as PubMed, the Cochrane Library, Embase, Scopus, and Web of Science. Results: It highlights HRW's effects on various aspects of athletic performance, including endurance, strength, sprint times, lunge movements, countermovement jump height, and time to exhaustion. While the precise mechanisms by which HRW affects athletic performance remain unclear, this review investigates its general molecular biological mechanisms beyond the specific context of sports. This provides a theoretical foundation for future research aimed at understanding how HRW can enhance athletic performance. HRW targets the harmful reactive oxygen and nitrogen species produced during intense exercise, thereby reducing oxidative stress-a critical factor in muscle fatigue, inflammation, and diminished athletic performance. HRW helps to scavenge hydroxyl radicals and peroxynitrite, regulate antioxidant enzymes, mitigate lipid peroxidation, reduce inflammation, protect against mitochondrial dysfunction, and modulate cellular signaling pathways. Conclusions: In summary, while a few studies have indicated that HRW may not produce significant beneficial effects, the majority of research supports the conclusion that HRW may enhance athletic performance across various sports. The potential mechanisms underlying these benefits are thought to involve HRW's role as a selective antioxidant, its impact on oxidative stress, and its regulation of redox homeostasis. However, the specific molecular biological mechanisms through which HRW improves athletic performance remain to be fully elucidated.
Collapse
Affiliation(s)
- Qiaorui Zhou
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Huixin Li
- School of Sport Science, Beijing Sport University, Beijing 100084, China;
| | - Ye Zhang
- Sport Coaching College, Beijing Sport University, Beijing 100084, China;
| | - Yirui Zhao
- China Ice and Snow Sports College, Beijing Sport University, Beijing 100084, China;
| | - Can Wang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Chang Liu
- School of Sport Science, Beijing Sport University, Beijing 100084, China;
| |
Collapse
|
27
|
Zhang D, Feng F, Chen Y, Sui J, Ding L. The potential of marine natural products and their synthetic derivatives as drugs targeting ion channels. Eur J Med Chem 2024; 276:116644. [PMID: 38971051 DOI: 10.1016/j.ejmech.2024.116644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/08/2024]
Abstract
Ion channels are a type of protein channel that play a vital role in numerous physiological functions by facilitating the passage of ions through cell membranes, thereby enabling ion and electrical signal transmission. As a crucial target for drug action, ion channels have been implicated in various diseases. Many natural products from marine organisms, such as fungi, algae, sponges, and sea cucumber, etc. have been found to have activities related to ion channels for decades. These interesting natural product molecules undoubtedly bring good news for the treatment of neurological and cardiovascular diseases. In this review, 92 marine natural products and their synthetic derivatives with ion channel-related activities that were identified during the period 2000-2024 were systematically reviewed. The synthesis and mechanisms of action of selected compounds were also discussed, aiming to offer insights for the development of drugs targeting ion channels.
Collapse
Affiliation(s)
- Dashuai Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, School of Pharmacy, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Fangjian Feng
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, School of Pharmacy, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yaoyao Chen
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, School of Pharmacy, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jingyao Sui
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, School of Pharmacy, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lijian Ding
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, School of Pharmacy, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
28
|
Ferreira MDA, Lückemeyer DD, Martins F, Schran RG, da Silva AM, Gambeta E, Zamponi GW, Ferreira J. Pronociceptive role of spinal Ca v2.3 (R-type) calcium channels in a mouse model of postoperative pain. Br J Pharmacol 2024; 181:3594-3609. [PMID: 38812100 DOI: 10.1111/bph.16407] [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: 08/07/2023] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND More than 80% of patients may experience acute pain after a surgical procedure, and this is often refractory to pharmacological intervention. The identification of new targets to treat postoperative pain is necessary. There is an association of polymorphisms in the Cav2.3 gene with postoperative pain and opioid consumption. Our study aimed to identify Cav2.3 as a potential target to treat postoperative pain and to reduce opioid-related side effects. EXPERIMENTAL APPROACH A plantar incision model was established in adult male and female C57BL/6 mice. Cav2.3 expression was detected by qPCR and suppressed by siRNA treatment. The antinociceptive efficacy and safety of a Cav2.3 blocker-alone or together with morphine-was also assessed after surgery. KEY RESULTS Paw incision in female and male mice caused acute nociception and increased Cav2.3 mRNA expression in the spinal cord but not in the incised tissue. Intrathecal treatment with siRNA against Cav2.3, but not with a scrambled siRNA, prevented the development of surgery-induced nociception in both male and female mice, with female mice experiencing long-lasting effects. High doses of i.t. SNX-482, a Cav2.3 channel blocker, or morphine injected alone, reversed postoperative nociception but also induced side effects. A combination of lower doses of morphine and SNX-482 mediated a long-lasting reversal of postsurgical pain in female and male mice. CONCLUSION Our results demonstrate that Cav2.3 has a pronociceptive role in the induction of postoperative pain, indicating that it is a potential target for the development of therapeutic approaches for the treatment of postoperative pain.
Collapse
Affiliation(s)
- Marcella de Amorim Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Debora Denardin Lückemeyer
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Anesthesiology, Pain Research Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fernanda Martins
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Roberta Giusti Schran
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Ana Merian da Silva
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Eder Gambeta
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Juliano Ferreira
- Graduate Program in Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
29
|
Hwang Y, Park JH, Kim HC, Shin EJ. Nimodipine attenuates neuroinflammation and delayed apoptotic neuronal death induced by trimethyltin in the dentate gyrus of mice. J Mol Histol 2024; 55:721-740. [PMID: 39083161 DOI: 10.1007/s10735-024-10226-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/12/2024] [Indexed: 10/10/2024]
Abstract
L-type voltage-gated calcium channels (L-VGCCs) are thought to be involved in epileptogenesis and acute excitotoxicity. However, little is known about the role of L-VGCCs in neuroinflammation or delayed neuronal death following excitotoxic insult. We examined the effects of repeated treatment with the L-VGCC blocker nimodipine on neuroinflammatory changes and delayed neuronal apoptosis in the dentate gyrus following trimethyltin (TMT)-induced convulsions. Male C57BL/6 N mice were administered TMT (2.6 mg/kg, i.p.), and the expression of the Cav1.2 and Cav1.3 subunits of L-VGCC were evaluated. The expression of both subunits was significantly decreased; however, the astroglial expression of Cav1.3 L-VGCC was significantly induced at 6 and 10 days after TMT treatment. Furthermore, astroglial Cav1.3 L-VGCCs colocalized with both the pro-inflammatory phenotype marker C3 and the anti-inflammatory phenotype marker S100A10 of astrocytes. Nimodipine (5 mg/kg, i.p. × 5 at 12-h intervals) did not significantly affect TMT-induced astroglial activation. However, nimodipine significantly attenuated the pro-inflammatory phenotype changes, while enhancing the anti-inflammatory phenotype changes in astrocytes after TMT treatment. Consistently, nimodipine reduced the levels of pro-inflammatory astrocytes-to-microglia mediators, while increasing the levels of anti-inflammatory astrocytes-to-microglia mediators. These effects were accompanied by an increase in the phosphorylation of extracellular signal-regulated kinase (ERK), supporting our previous finding that p-ERK is a signaling factor that regulates astroglial phenotype changes. In addition, nimodipine significantly attenuated TMT-induced microglial activation and delayed apoptosis of dentate granule neurons. Our results suggest that L-VGCC blockade attenuates neuroinflammation and delayed neurotoxicity following TMT-induced convulsions through the regulation of astroglial phenotypic changes by promoting ERK signaling.
Collapse
Affiliation(s)
- Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jung Hoon Park
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
30
|
Kovács Z, Rauch E, D’Agostino DP, Ari C. Putative Role of Adenosine A1 Receptors in Exogenous Ketone Supplements-Evoked Anti-Epileptic Effect. Int J Mol Sci 2024; 25:9869. [PMID: 39337356 PMCID: PMC11432942 DOI: 10.3390/ijms25189869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Approximately 30% of patients with epilepsy are drug-refractory. There is an urgent need to elucidate the exact pathophysiology of different types of epilepsies and the mechanisms of action of both antiseizure medication and metabolic therapies to treat patients more effectively and safely. For example, it has been demonstrated that exogenous ketone supplement (EKS)-generated therapeutic ketosis, as a metabolic therapy, may decrease epileptic activity in both animal models and humans, but its exact mechanism of action is unknown. However, it was demonstrated that therapeutic ketosis, among others, can increase adenosine level, which may enhance activity of A1 adenosine receptors (A1Rs) in the brain. It has also been demonstrated previously that adenosine has anti-epileptic effect through A1Rs in different models of epilepsies. Thus, it is possible that (i) therapeutic ketosis generated by the administration of EKSs may exert its anti-epileptic effect through, among other mechanisms, increased adenosine level and A1R activity and that (ii) the enhanced activity of A1Rs may be a necessary anti-epileptic mechanism evoked by EKS administration-generated ketosis. Moreover, EKSs can evoke and maintain ketosis without severe side effects. These results also suggest that the therapeutic application of EKS-generated ketosis may be a promising opportunity to treat different types of epilepsies. In this literature review, we specifically focus on the putative role of A1Rs in the anti-epileptic effect of EKS-induced ketosis.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Biology, BDTTC, ELTE Eötvös Loránd University, Károlyi Gáspár tér 4., 9700 Szombathely, Hungary or (Z.K.); (E.R.)
| | - Enikő Rauch
- Department of Biology, BDTTC, ELTE Eötvös Loránd University, Károlyi Gáspár tér 4., 9700 Szombathely, Hungary or (Z.K.); (E.R.)
- Institute of Biology, University of Pécs, Ifjúság Str. 6, 7624 Pécs, Hungary
| | - Dominic P. D’Agostino
- Ketone Technologies LLC., Tampa, FL 33612, USA;
- Department of Molecular Pharmacology and Physiology, Laboratory of Metabolic Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Institute for Human and Machine Cognition, Ocala, FL 34471, USA
| | - Csilla Ari
- Ketone Technologies LLC., Tampa, FL 33612, USA;
- Department of Psychology, Behavioral Neuroscience Research Laboratory, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
31
|
Saha O, Melo de Farias AR, Pelletier A, Siedlecki-Wullich D, Landeira BS, Gadaut J, Carrier A, Vreulx AC, Guyot K, Shen Y, Bonnefond A, Amouyel P, Tcw J, Kilinc D, Queiroz CM, Delahaye F, Lambert JC, Costa MR. The Alzheimer's disease risk gene BIN1 regulates activity-dependent gene expression in human-induced glutamatergic neurons. Mol Psychiatry 2024; 29:2634-2646. [PMID: 38514804 PMCID: PMC11420064 DOI: 10.1038/s41380-024-02502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
Bridging Integrator 1 (BIN1) is the second most important Alzheimer's disease (AD) risk gene, but its physiological roles in neurons and its contribution to brain pathology remain largely elusive. In this work, we show that BIN1 plays a critical role in the regulation of calcium homeostasis, electrical activity, and gene expression of glutamatergic neurons. Using single-cell RNA-sequencing on cerebral organoids generated from isogenic BIN1 wild type (WT), heterozygous (HET) and homozygous knockout (KO) human-induced pluripotent stem cells (hiPSCs), we show that BIN1 is mainly expressed by oligodendrocytes and glutamatergic neurons, like in the human brain. Both BIN1 HET and KO cerebral organoids show specific transcriptional alterations, mainly associated with ion transport and synapses in glutamatergic neurons. We then demonstrate that BIN1 cell-autonomously regulates gene expression in glutamatergic neurons by using a novel protocol to generate pure culture of hiPSC-derived induced neurons (hiNs). Using this system, we also show that BIN1 plays a key role in the regulation of neuronal calcium transients and electrical activity via its interaction with the L-type voltage-gated calcium channel Cav1.2. BIN1 KO hiNs show reduced activity-dependent internalization and higher Cav1.2 expression compared to WT hiNs. Pharmacological blocking of this channel with clinically relevant doses of nifedipine, a calcium channel blocker, partly rescues electrical and gene expression alterations in BIN1 KO glutamatergic neurons. Further, we show that transcriptional alterations in BIN1 KO hiNs that affect biological processes related to calcium homeostasis are also present in glutamatergic neurons of the human brain at late stages of AD pathology. Together, these findings suggest that BIN1-dependent alterations in neuronal properties could contribute to AD pathophysiology and that treatment with low doses of clinically approved calcium blockers should be considered as an option to slow disease-onset and progression.
Collapse
Affiliation(s)
- Orthis Saha
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Ana Raquel Melo de Farias
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Alexandre Pelletier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Dolores Siedlecki-Wullich
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Bruna Soares Landeira
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Johanna Gadaut
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Arnaud Carrier
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Anaïs-Camille Vreulx
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Karine Guyot
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Yun Shen
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Amelie Bonnefond
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Philippe Amouyel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Julia Tcw
- Department of Pharmacology, Physiology & Biophysics, Boston University, Chobanian & Avedisian School of Medicine, Boston, MA, 02118, USA
- Bioinformatics Program, Faculty of Computing & Data Sciences, Boston University, Boston, MA, 02115, USA
| | - Devrim Kilinc
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Claudio Marcos Queiroz
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil
| | - Fabien Delahaye
- Univ. Lille, Inserm, CNRS, CHU Lille, Institut Pasteur de Lille, U1283-UMR 8199 EGID, Pôle Recherche, 1 Place de Verdun, 59045, Lille, Cedex, France
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France
| | - Marcos R Costa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, DISTALZ, 1 rue du Professeur Calmette, 59019, Lille, France.
- Brain Institute, Federal University of Rio Grande do Norte, Av. Senador Salgado Filho, 3000, Campus Universitário, Lagoa, Nova, 59078-970, Natal, Brazil.
| |
Collapse
|
32
|
Naseri S, Samaram H, Naghavi N, Rassouli MB, Mousavinezhad M. Types of Short-Duration Electrical Stimulation-Induced Efficiency in the Axonal Regeneration and Recovery: Comparative in Vivo Study in Rat Model of Repaired Sciatic Nerve and its Tibial Branch after Transection Injury. Neurochem Res 2024; 49:2469-2479. [PMID: 38856888 DOI: 10.1007/s11064-024-04154-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/25/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
The restoration of adequate function and sensation in nerves following an injury is often insufficient. Electrical stimulation (ES) applied during nerve repair can promote axon regeneration, which may enhance the likelihood of successful functional recovery. However, increasing operation time and complexity are associated with limited clinical use of ES. This study aims to better assess whether short-duration ES types (voltage mode vs. current mode) are able to produce enhanced regenerative activity following peripheral nerve repair in rat models. Wistar rats were randomly divided into 3 groups: no ES (control), 30-minute ES with a current pulse, and 30-minute ES with a voltage pulse. All groups underwent sciatic nerve transection and repair using a silicone tube to bridge the 6-mm gap between the stumps. In the 2 groups other than the control, ES was applied after the surgical repair. Outcomes were evaluated using electrophysiology, histology, and serial walking track analysis. Biweekly walking tracks test over 12 weeks revealed that subjects that underwent ES experienced more rapid functional improvement than subjects that underwent repair alone. Electrophysiological analysis of the newly intratubular sciatic nerve at week 12 revealed strong motor function recovery in rats that underwent 30-minute ES. Histologic analysis of the sciatic nerve and its tibial branch at 12 weeks demonstrated robust axon regrowth in all groups. Both types of short-duration ES applied during nerve repair can promote axon regrowth and enhance the chances of successful functional recovery.
Collapse
Affiliation(s)
- Sareh Naseri
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran
| | - Hosein Samaram
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran
| | - Nadia Naghavi
- Electrical Engineering Department, Faculty of Engineering, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Razavi Khorasan Province, 9177948374, Iran.
| | | | - Maryam Mousavinezhad
- Biology Department, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
33
|
Ahmadi S, Majidi M, Koraei M, Vasef S. The Inflammation/NF-κB and BDNF/TrkB/CREB Pathways in the Cerebellum Are Implicated in the Changes in Spatial Working Memory After Both Morphine Dependence and Withdrawal in Rat. Mol Neurobiol 2024; 61:6721-6733. [PMID: 38347284 DOI: 10.1007/s12035-024-03993-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/27/2024] [Indexed: 08/22/2024]
Abstract
We aimed to explore the impact of the cerebellum on the decline in spatial working memory following morphine dependence and withdrawal. Two groups of male Wistar rats received intraperitoneal injections of either saline (1 ml/kg) or morphine (10 mg/kg) twice daily for 10 days, serving as the control and dependent groups. Additionally, a withdrawal group underwent a 30-day withdrawal period after the dependence phase. Spatial working memory was assessed using a Y maze test. ELISA and western blot were used to assess protein levels in the cerebellum. On day 1, morphine impaired spatial working memory, deteriorated further after 10 days of morphine use, and nearly returned to its initial level following a 30-day withdrawal period. On day 10, significant increases in TNF-α, IL-1β, and CXCL12 and a notable decrease in IL-10 levels were detected in the morphine-dependent group, which did not completely restore in the withdrawal group. The protein levels of CXCR4, TLR4, P2X7R, and NF-κB sharply increased in the morphine-dependent group. However, these levels almost returned to normal after withdrawal. In the morphine-dependent group, BDNF decreased, while TrkB and CREB1 increased noticeably. Nevertheless, after withdrawal, TrkB and CREB1 but not BDNF levels returned to normal. In the morphine-dependent group, both CACNA1 and KCNMA1 decreased significantly and after withdrawal, only KCNMA1 showed partial restoration, while CACNA1 did not. It can be concluded that inflammation/NF-κB and BDNF/TrkB/CREB pathways play key roles in neural adaptation within the cerebellum, contributing to the decline in spatial working memory after both morphine dependence and withdrawal.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran.
| | - Mohammad Majidi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Maryam Koraei
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Samira Vasef
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| |
Collapse
|
34
|
Egido-Betancourt HX, Strowd III RE, Raab-Graham KF. Potential roles of voltage-gated ion channel disruption in Tuberous Sclerosis Complex. Front Mol Neurosci 2024; 17:1404884. [PMID: 39253727 PMCID: PMC11381416 DOI: 10.3389/fnmol.2024.1404884] [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: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 09/11/2024] Open
Abstract
Tuberous Sclerosis Complex (TSC) is a lynchpin disorder, as it results in overactive mammalian target of rapamycin (mTOR) signaling, which has been implicated in a multitude of disease states. TSC is an autosomal dominant disease where 90% of affected individuals develop epilepsy. Epilepsy results from aberrant neuronal excitability that leads to recurring seizures. Under neurotypical conditions, the coordinated activity of voltage-gated ion channels keep neurons operating in an optimal range, thus providing network stability. Interestingly, loss or gain of function mutations in voltage-gated potassium, sodium, or calcium channels leads to altered excitability and seizures. To date, little is known about voltage-gated ion channel expression and function in TSC. However, data is beginning to emerge on how mTOR signaling regulates voltage-gated ion channel expression in neurons. Herein, we provide a comprehensive review of the literature describing common seizure types in patients with TSC, and suggest possible parallels between acquired epilepsies with known voltage-gated ion channel dysfunction. Furthermore, we discuss possible links toward mTOR regulation of voltage-gated ion channels expression and channel kinetics and the underlying epileptic manifestations in patients with TSC.
Collapse
Affiliation(s)
- Hailey X. Egido-Betancourt
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Roy E. Strowd III
- Department of Neurology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kimberly F. Raab-Graham
- Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
35
|
Jin X, Huang J, Wang H, Wang K, Yan N. A versatile residue numbering scheme for Na v and Ca v channels. Cell Chem Biol 2024; 31:1394-1404. [PMID: 39151406 DOI: 10.1016/j.chembiol.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/01/2024] [Accepted: 07/18/2024] [Indexed: 08/19/2024]
Abstract
Voltage-gated sodium (Nav) and calcium (Cav) channels are responsible for the initiation of electrical signals. They have long been targeted for the treatment of various diseases. The mounting number of cryoelectron microscopy (cryo-EM) structures for diverse subtypes of Nav and Cav channels from multiple organisms necessitates a generic residue numbering system to establish the structure-function relationship and to aid rational drug design or optimization. Here we suggest a structure-based residue numbering scheme, centering around the most conserved residues on each of the functional segments. We elaborate the generic numbers through illustrative examples, focusing on representative drug-binding sites of eukaryotic Nav and Cav channels. We also extend the numbering scheme to compare common disease mutations among different Nav subtypes. Application of the generic residue numbering scheme affords immediate insights into hotspots for pathogenic mutations and critical loci for drug binding and will facilitate drug discovery targeting Nav and Cav channels.
Collapse
Affiliation(s)
- Xueqin Jin
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Jian Huang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Huan Wang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kan Wang
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation, Guangming District, Shenzhen, Guangdong Province 518107, China; Shenzhen Bay Laboratory, Guangming District, Shenzhen, Guangdong Province 518132, China.
| |
Collapse
|
36
|
Di Micco V, Affronte L, Khinchi MS, Rønde G, Miranda MJ, Hammer TB, Specchio N, Beniczky S, Olofsson K, Møller RS, Gardella E. Seizure and movement disorder in CACNA1E developmental and epileptic encephalopathy: Two sides of the same coin or same side of two different coins? Epileptic Disord 2024; 26:520-526. [PMID: 38780451 DOI: 10.1002/epd2.20242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 04/23/2024] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
Pathogenic variants in CACNA1E are associated with early-onset epileptic and developmental encephalopathy (DEE). Severe to profound global developmental delay, early-onset refractory seizures, severe hypotonia, and macrocephaly are the main clinical features. Patients harboring the recurrent CACNA1E variant p.(Gly352Arg) typically present with the combination of early-onset DEE, dystonia/dyskinesia, and contractures. We describe a 2-year-and-11-month-old girl carrying the p.(Gly352Arg) CACNA1E variant. She has a severe DEE with very frequent drug-resistant seizures, profound hypotonia, and episodes of dystonia and dyskinesia. Long-term video-EEG-monitoring documented subsequent tonic asymmetric seizures during wakefulness and mild paroxysmal dyskinesias of the trunk out of sleep which were thought to be a movement disorder and instead turned out to be focal hyperkinetic seizures. This is the first documented description of the EEG findings in this disorder. Our report highlights a possible overlap between cortical and subcortical phenomena in CACNA1E-DEE. We also underline how a careful electro-clinical evaluation might be necessary for a correct discernment between the two disorders, playing a fundamental role in the clinical assessment and proper management of children with CACNA1E-DEE.
Collapse
Affiliation(s)
- Valentina Di Micco
- Department of Clinical Neurophysiology, Danish Epilepsy Centre, Dianalund, Denmark
- Epilepsy and Movement Disorders, Neurology Unit, Bambino Gesù Children's Hospital, Rome, Italy
- Member of the European Reference Network EpiCARE
| | - Leonardo Affronte
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
- Child Neuropsychiatry, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | | | - Gitte Rønde
- Department of Pediatrics, Herlev and Gentofte University Hospital, Copenhagen University, Copenhagen, Denmark
| | - Maria Jose Miranda
- Department of Pediatrics, Herlev and Gentofte University Hospital, Copenhagen University, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Trine Bjørg Hammer
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
| | - Nicola Specchio
- Epilepsy and Movement Disorders, Neurology Unit, Bambino Gesù Children's Hospital, Rome, Italy
- Member of the European Reference Network EpiCARE
| | - Sándor Beniczky
- Department of Clinical Neurophysiology, Danish Epilepsy Centre, Dianalund, Denmark
- Member of the European Reference Network EpiCARE
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
| | - Kern Olofsson
- Department of Child Neurology, Danish Epilepsy Centre, Dianalund, Denmark
| | - Rikke S Møller
- Member of the European Reference Network EpiCARE
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
- Department of Clinical Genetics and Precision Treatment, Danish Epilepsy Centre, Dianalund, Denmark
| | - Elena Gardella
- Department of Clinical Neurophysiology, Danish Epilepsy Centre, Dianalund, Denmark
- Member of the European Reference Network EpiCARE
- Department of Epilepsy Genetics and Personalized Medicine, Danish Epilepsy Centre, Dianalund, Denmark
- Department of Clinical Genetics and Precision Treatment, Danish Epilepsy Centre, Dianalund, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
37
|
Chichorro JG, Gambeta E, Baggio DF, Zamponi GW. Voltage-gated Calcium Channels as Potential Therapeutic Targets in Migraine. THE JOURNAL OF PAIN 2024; 25:104514. [PMID: 38522594 DOI: 10.1016/j.jpain.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Migraine is a complex and highly incapacitating neurological disorder that affects around 15% of the general population with greater incidence in women, often at the most productive age of life. Migraine physiopathology is still not fully understood, but it involves multiple mediators and events in the trigeminovascular system and the central nervous system. The identification of calcitonin gene-related peptide as a key mediator in migraine physiopathology has led to the development of effective and highly selective antimigraine therapies. However, this treatment is neither accessible nor effective for all migraine sufferers. Thus, a better understanding of migraine mechanisms and the identification of potential targets are still clearly warranted. Voltage-gated calcium channels (VGCCs) are widely distributed in the trigeminovascular system, and there is accumulating evidence of their contribution to the mechanisms associated with headache pain. Several drugs used in migraine abortive or prophylactic treatment target VGCCs, which probably contributes to their analgesic effect. This review aims to summarize the current evidence of VGGC contribution to migraine physiopathology and to discuss how current pharmacological options for migraine treatment interfere with VGGC function. PERSPECTIVE: Calcitonin gene-related peptide (CGRP) represents a major migraine mediator, but few studies have investigated the relationship between CGRP and VGCCs. CGRP release is calcium channel-dependent and VGGCs are key players in familial migraine. Further studies are needed to determine whether VGCCs are suitable molecular targets for treating migraine.
Collapse
Affiliation(s)
- Juliana G Chichorro
- Biological Sciences Sector, Department of Pharmacology, Federal University of Parana, Curitiba, Parana, Brazil.
| | - Eder Gambeta
- Cumming School of Medicine, Department of Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Darciane F Baggio
- Biological Sciences Sector, Department of Pharmacology, Federal University of Parana, Curitiba, Parana, Brazil
| | - Gerald W Zamponi
- Cumming School of Medicine, Department of Clinical Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
38
|
Chin M, Kaeser PS. The intracellular C-terminus confers compartment-specific targeting of voltage-gated calcium channels. Cell Rep 2024; 43:114428. [PMID: 38996073 PMCID: PMC11441329 DOI: 10.1016/j.celrep.2024.114428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
To achieve the functional polarization that underlies brain computation, neurons sort protein material into distinct compartments. Ion channel composition, for example, differs between axons and dendrites, but the molecular determinants for their polarized trafficking remain obscure. Here, we identify mechanisms that target voltage-gated Ca2+ channels (CaVs) to distinct subcellular compartments. In hippocampal neurons, CaV2s trigger neurotransmitter release at the presynaptic active zone, and CaV1s localize somatodendritically. After knockout of all three CaV2s, expression of CaV2.1, but not CaV1.3, restores neurotransmitter release. We find that chimeric CaV1.3s with CaV2.1 intracellular C-termini localize to the active zone, mediate synaptic vesicle exocytosis, and render release sensitive to CaV1 blockers. This dominant targeting function of the CaV2.1 C-terminus requires the first EF hand in its proximal segment, and replacement of the CaV2.1 C-terminus with that of CaV1.3 abolishes CaV2.1 active zone localization and function. We conclude that CaV intracellular C-termini mediate compartment-specific targeting.
Collapse
Affiliation(s)
- Morven Chin
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
39
|
Paquin-Lefebvre F, Holcman D. Voltage mapping in subcellular nanodomains using electro-diffusion modeling. J Chem Phys 2024; 161:034108. [PMID: 39007374 DOI: 10.1063/5.0215900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Voltage distribution in sub-cellular micro-domains such as neuronal synapses, small protrusions, or dendritic spines regulates the opening and closing of ionic channels, energy production, and thus, cellular homeostasis and excitability. Yet how voltage changes at such a small scale in vivo remains challenging due to the experimental diffraction limit, large signal fluctuations, and the still limited resolution of fast voltage indicators. Here, we study the voltage distribution in nano-compartments using a computational approach based on the Poisson-Nernst-Planck equations for the electro-diffusion motion of ions, where inward and outward fluxes are generated between channels. We report a current-voltage (I-V) logarithmic relationship generalizing Nernst law that reveals how the local membrane curvature modulates the voltage. We further find that an influx current penetrating a cellular electrolyte can lead to perturbations from tens to hundreds of nanometers deep, depending on the local channel organization. Finally, we show that the neck resistance of dendritic spines can be completely shunted by the transporters located on the head boundary, facilitating ionic flow. To conclude, we propose that voltage is regulated at a subcellular level by channel organization, membrane curvature, and narrow passages.
Collapse
Affiliation(s)
- Frédéric Paquin-Lefebvre
- Group of Data Modeling, Computational Biology and Applied Mathematics, École Normale Supérieure - Université PSL, 75005 Paris, France
| | - David Holcman
- Group of Data Modeling, Computational Biology and Applied Mathematics, École Normale Supérieure - Université PSL, 75005 Paris, France
- Department of Applied Mathematics and Theoretical Physics and Churchill College, University of Cambridge, Cambridge CB3 0WA, United Kingdom
| |
Collapse
|
40
|
Karagianni K, Dafou D, Xanthopoulos K, Sklaviadis T, Kanata E. RNA editing regulates glutamatergic synapses in the frontal cortex of a molecular subtype of Amyotrophic Lateral Sclerosis. Mol Med 2024; 30:101. [PMID: 38997636 PMCID: PMC11241978 DOI: 10.1186/s10020-024-00863-2] [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: 04/04/2024] [Accepted: 06/12/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Amyotrophic Lateral Sclerosis (ALS) is a highly heterogenous neurodegenerative disorder that primarily affects upper and lower motor neurons, affecting additional cell types and brain regions. Underlying molecular mechanisms are still elusive, in part due to disease heterogeneity. Molecular disease subtyping through integrative analyses including RNA editing profiling is a novel approach for identification of molecular networks involved in pathogenesis. METHODS We aimed to highlight the role of RNA editing in ALS, focusing on the frontal cortex and the prevalent molecular disease subtype (ALS-Ox), previously determined by transcriptomic profile stratification. We established global RNA editing (editome) and gene expression (transcriptome) profiles in control and ALS-Ox cases, utilizing publicly available RNA-seq data (GSE153960) and an in-house analysis pipeline. Functional annotation and pathway analyses identified molecular processes affected by RNA editing alterations. Pearson correlation analyses assessed RNA editing effects on expression. Similar analyses on additional ALS-Ox and control samples (GSE124439) were performed for verification. Targeted re-sequencing and qRT-PCR analysis targeting CACNA1C, were performed using frontal cortex tissue from ALS and control samples (n = 3 samples/group). RESULTS We identified reduced global RNA editing in the frontal cortex of ALS-Ox cases. Differentially edited transcripts are enriched in synapses, particularly in the glutamatergic synapse pathway. Bioinformatic analyses on additional ALS-Ox and control RNA-seq data verified these findings. We identified increased recoding at the Q621R site in the GRIK2 transcript and determined positive correlations between RNA editing and gene expression alterations in ionotropic receptor subunits GRIA2, GRIA3 and the CACNA1C transcript, which encodes the pore forming subunit of a post-synaptic L-type calcium channel. Experimental data verified RNA editing alterations and editing-expression correlation in CACNA1C, highlighting CACNA1C as a target for further study. CONCLUSIONS We provide evidence on the involvement of RNA editing in the frontal cortex of an ALS molecular subtype, highlighting a modulatory role mediated though recoding and gene expression regulation on glutamatergic synapse related transcripts. We report RNA editing effects in disease-related transcripts and validated editing alterations in CACNA1C. Our study provides targets for further functional studies that could shed light in underlying disease mechanisms enabling novel therapeutic approaches.
Collapse
Affiliation(s)
- Korina Karagianni
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece
| | - Dimitra Dafou
- Department of Genetics, Development, and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 541 24, Thessaloniki, Greece
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, 57001, Thermi, Greece
| | - Theodoros Sklaviadis
- Laboratory of Pharmacology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Eirini Kanata
- Laboratory of Pharmacology, Department of Pharmacy, School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
41
|
Li X, Feng R, Guo Z, Meng Y, Zou Y, Liao W, Peng Q, Zhong H, Zhao W. Direct investigations of the effects of nicardipine on calcium channels of astrocytes by Atomic Force Microscopy. Talanta 2024; 274:125947. [PMID: 38537353 DOI: 10.1016/j.talanta.2024.125947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 05/04/2024]
Abstract
Calcium channel blockers (CCB) of astrocytes can blockade the calcium ions entry through the voltage gated calcium channels (VGCC), and is widely used in the diseases related with VGCC of astrocytes. But many aspects of the interaction mechanisms between the CCB and VGCC of astrocytes still remain unclear due to the limited resolution of the approaches. Herein the effects of the nicardipine (a type of CCB) on VGCC of astrocytes were investigated at very high spatial, force and electrical resolution by multiple modes of Atomic Force Microscopy (AFM) directly. The results reveal that after the addition of nicardipine, the recognition signals of VGCC disappeared; the specific unbinding forces vanished; the conductivity of the astrocytes decreased (the current decreased about 2.9 pA and the capacitance was doubled); the surface potential of the astrocytes reduced about 14.2 mV. The results of electrical properties investigations are consistent with the simulation experiments. The relations between these biophysical and biochemical properties of VGCC have been discussed. All these demonstrate that the interactions between nicardipine and VGCC have been studied at nanometer spatial resolution, at picoNewton force resolution and very high electrical signal resolution (pA in current, pF in capacitance and 0.1 mV in surface potential) level. The approaches are considered to be high resolution and high sensitivity, and will be helpful and useful in the further investigations of the effects of other types of CCB on ion channels, and will also be helpful in the investigations of mechanisms and therapy of ion channelopathies.
Collapse
Affiliation(s)
- Xinyu Li
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Rongrong Feng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Zeling Guo
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Yu Meng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Yulan Zou
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Wenchao Liao
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Qianwei Peng
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, People's Republic of China
| | - Haijian Zhong
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China.
| | - Weidong Zhao
- Key Laboratory of Biomaterials and Biofabrication in Tissue Engineering of Jiangxi Province, Gannan Medical University, Ganzhou, 341000, People's Republic of China; School of Medical Information Engineering, Gannan Medical University, Ganzhou, 341000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, People's Republic of China.
| |
Collapse
|
42
|
Currim F, Tanwar R, Brown-Leung JM, Paranjape N, Liu J, Sanders LH, Doorn JA, Cannon JR. Selective dopaminergic neurotoxicity modulated by inherent cell-type specific neurobiology. Neurotoxicology 2024; 103:266-287. [PMID: 38964509 PMCID: PMC11288778 DOI: 10.1016/j.neuro.2024.06.016] [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: 02/18/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative disease affecting millions of individuals worldwide. Hallmark features of PD pathology are the formation of Lewy bodies in neuromelanin-containing dopaminergic (DAergic) neurons of the substantia nigra pars compacta (SNpc), and the subsequent irreversible death of these neurons. Although genetic risk factors have been identified, around 90 % of PD cases are sporadic and likely caused by environmental exposures and gene-environment interaction. Mechanistic studies have identified a variety of chemical PD risk factors. PD neuropathology occurs throughout the brain and peripheral nervous system, but it is the loss of DAergic neurons in the SNpc that produce many of the cardinal motor symptoms. Toxicology studies have found specifically the DAergic neuron population of the SNpc exhibit heightened sensitivity to highly variable chemical insults (both in terms of chemical structure and mechanism of neurotoxic action). Thus, it has become clear that the inherent neurobiology of nigral DAergic neurons likely underlies much of this neurotoxic response to broad insults. This review focuses on inherent neurobiology of nigral DAergic neurons and how such neurobiology impacts the primary mechanism of neurotoxicity. While interactions with a variety of other cell types are important in disease pathogenesis, understanding how inherent DAergic biology contributes to selective sensitivity and primary mechanisms of neurotoxicity is critical to advancing the field. Specifically, key biological features of DAergic neurons that increase neurotoxicant susceptibility.
Collapse
Affiliation(s)
- Fatema Currim
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Reeya Tanwar
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Josephine M Brown-Leung
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA
| | - Neha Paranjape
- Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Jennifer Liu
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Laurie H Sanders
- Departments of Neurology and Pathology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Center for Neurodegeneration and Neurotherapeutics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jonathan A Doorn
- Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, IN 47901, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47901, USA.
| |
Collapse
|
43
|
Posa A, Kornhuber M. P/Q-type calcium channel antibody-associated headache disorder (P/Q-HaND). Pain Pract 2024; 24:871-872. [PMID: 38450891 DOI: 10.1111/papr.13366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Affiliation(s)
- Andreas Posa
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Malte Kornhuber
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
- Department of Neurology, Helios Hospital Sangerhausen, Sangerhausen, Germany
| |
Collapse
|
44
|
Kim JW, Yong AJH, Aisenberg EE, Lobel JH, Wang W, Dawson TM, Dawson VL, Gao R, Jan YN, Bateup HS, Ingolia NT. Molecular recording of calcium signals via calcium-dependent proximity labeling. Nat Chem Biol 2024; 20:894-905. [PMID: 38658655 DOI: 10.1038/s41589-024-01603-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 03/08/2024] [Indexed: 04/26/2024]
Abstract
Calcium ions serve as key intracellular signals. Local, transient increases in calcium concentrations can activate calcium sensor proteins that in turn trigger downstream effectors. In neurons, calcium transients play a central role in regulating neurotransmitter release and synaptic plasticity. However, it is challenging to capture the molecular events associated with these localized and ephemeral calcium signals. Here we present an engineered biotin ligase that generates permanent molecular traces in a calcium-dependent manner. The enzyme, calcium-dependent BioID (Cal-ID), biotinylates nearby proteins within minutes in response to elevated local calcium levels. The biotinylated proteins can be identified via mass spectrometry and visualized using microscopy. In neurons, Cal-ID labeling is triggered by neuronal activity, leading to prominent protein biotinylation that enables transcription-independent activity labeling in the brain. In summary, Cal-ID produces a biochemical record of calcium signals and neuronal activity with high spatial resolution and molecular specificity.
Collapse
Affiliation(s)
- J Wren Kim
- Department of Molecular and Cell Biology at the University of California, Berkeley, Berkeley, CA, USA
| | - Adeline J H Yong
- Department of Physiology at the University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute at the University of California, San Francisco, San Francisco, CA, USA
| | - Erin E Aisenberg
- Helen Wills Neuroscience Institute at the University of California, Berkeley, Berkeley, CA, USA
| | - Joseph H Lobel
- Department of Molecular and Cell Biology at the University of California, Berkeley, Berkeley, CA, USA
| | - Wei Wang
- Department of Chemistry at the University of Illinois, Chicago, Chicago, IL, USA
| | - Ted M Dawson
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruixuan Gao
- Department of Chemistry at the University of Illinois, Chicago, Chicago, IL, USA
| | - Yuh Nung Jan
- Department of Physiology at the University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute at the University of California, San Francisco, San Francisco, CA, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology at the University of California, Berkeley, Berkeley, CA, USA
- Helen Wills Neuroscience Institute at the University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Nicholas T Ingolia
- Department of Molecular and Cell Biology at the University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
45
|
Yu XY, Sun QM, Lu RP, Wei B, Wang XY, Pan LH. Clinical features and genetic analysis of developmental and epileptic encephalopathy caused by biallelic variants of CACNA1B. Heliyon 2024; 10:e32693. [PMID: 39005920 PMCID: PMC11239463 DOI: 10.1016/j.heliyon.2024.e32693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
Objective To analyze the clinical features and genetic etiology of a patient with developmental and epileptic encephalopathy. Methods The clinical information and peripheral blood of the patient and their family members were collected before the whole exome sequencing analysis was performed and Sanger sequencing was employed to verify the potential variant. Results The patient presented with epilepsy and cerebral palsy with his parents, brother, and sister being all healthy. Whole exome sequencing analysis revealed that the child carried the paternal c.823del (p. R275Gfs*31) heterozygous variant and the maternal c.2456del (p.V819Gfs*190) heterozygous variant of the CACNA1B gene. Pedigree verification found that the elder brother and amniotic fluid of fetus in womb carried the paternal c.823del heterozygous variant, and the elder sister carried the maternal c.2456del heterozygous variant, which conformed to the law of autosomal recessive inheritance. Neither of these two variants has been reported in the literature and has not been included in the Genomic Mutation Frequency Database (gnomAD); according to the American Academy of Medical Genetics and Genomics Variation Grading Guidelines (ACMG), both variants are classified as pathogenic variants (PVS1+PM2-Supporting + PM3). Conclusion This study reported the first case of a child with neurodevelopmental disorder and epilepsy caused by a new compound heterozygous variant of the CACNA1B gene in China, clarified its genetic etiology, enriched the mutation spectrum and disease spectrum of CACNA1B gene, and provided a basis for prenatal diagnosis of the family.
Collapse
Affiliation(s)
- Xin-you Yu
- Gerneral Hospital of Ningxia Medical University, Yin'chuan, Ningxia, 750004, China
| | - Qing-mei Sun
- Gansu Province Maternal and Child Health Care Hospital, Lan'zhou, Gansu, 730000, China
| | - Rui-ping Lu
- Gerneral Hospital of Ningxia Medical University, Yin'chuan, Ningxia, 750004, China
| | - Bo Wei
- Gerneral Hospital of Ningxia Medical University, Yin'chuan, Ningxia, 750004, China
| | - Xiao-yan Wang
- Gerneral Hospital of Ningxia Medical University, Yin'chuan, Ningxia, 750004, China
| | - Li-hua Pan
- Gerneral Hospital of Ningxia Medical University, Yin'chuan, Ningxia, 750004, China
| |
Collapse
|
46
|
Ng ACH, Chahine M, Scantlebury MH, Appendino JP. Channelopathies in epilepsy: an overview of clinical presentations, pathogenic mechanisms, and therapeutic insights. J Neurol 2024; 271:3063-3094. [PMID: 38607431 DOI: 10.1007/s00415-024-12352-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Pathogenic variants in genes encoding ion channels are causal for various pediatric and adult neurological conditions. In particular, several epilepsy syndromes have been identified to be caused by specific channelopathies. These encompass a spectrum from self-limited epilepsies to developmental and epileptic encephalopathies spanning genetic and acquired causes. Several of these channelopathies have exquisite responses to specific antiseizure medications (ASMs), while others ASMs may prove ineffective or even worsen seizures. Some channelopathies demonstrate phenotypic pleiotropy and can cause other neurological conditions outside of epilepsy. This review aims to provide a comprehensive exploration of the pathophysiology of seizure generation, ion channels implicated in epilepsy, and several genetic epilepsies due to ion channel dysfunction. We outline the clinical presentation, pathogenesis, and the current state of basic science and clinical research for these channelopathies. In addition, we briefly look at potential precision therapy approaches emerging for these disorders.
Collapse
Affiliation(s)
- Andy Cheuk-Him Ng
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Division of Neurology, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada
| | - Mohamed Chahine
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
- CERVO, Brain Research Centre, Quebec City, Canada
| | - Morris H Scantlebury
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada
- Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Calgary, Canada
| | - Juan P Appendino
- Clinical Neuroscience and Pediatric Neurology, Department of Pediatrics, Cumming School of Medicine, Alberta Children's Hospital, University of Calgary, 28 Oki Drive NW, Calgary, AB, T3B 6A8, Canada.
| |
Collapse
|
47
|
Pio-Lopez L, Levin M. Aging as a loss of morphostatic information: A developmental bioelectricity perspective. Ageing Res Rev 2024; 97:102310. [PMID: 38636560 DOI: 10.1016/j.arr.2024.102310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/21/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Maintaining order at the tissue level is crucial throughout the lifespan, as failure can lead to cancer and an accumulation of molecular and cellular disorders. Perhaps, the most consistent and pervasive result of these failures is aging, which is characterized by the progressive loss of function and decline in the ability to maintain anatomical homeostasis and reproduce. This leads to organ malfunction, diseases, and ultimately death. The traditional understanding of aging is that it is caused by the accumulation of molecular and cellular damage. In this article, we propose a complementary view of aging from the perspective of endogenous bioelectricity which has not yet been integrated into aging research. We propose a view of aging as a morphostasis defect, a loss of biophysical prepattern information, encoding anatomical setpoints used for dynamic tissue and organ homeostasis. We hypothesize that this is specifically driven by abrogation of the endogenous bioelectric signaling that normally harnesses individual cell behaviors toward the creation and upkeep of complex multicellular structures in vivo. Herein, we first describe bioelectricity as the physiological software of life, and then identify and discuss the links between bioelectricity and life extension strategies and age-related diseases. We develop a bridge between aging and regeneration via bioelectric signaling that suggests a research program for healthful longevity via morphoceuticals. Finally, we discuss the broader implications of the homologies between development, aging, cancer and regeneration and how morphoceuticals can be developed for aging.
Collapse
Affiliation(s)
- Léo Pio-Lopez
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA
| | - Michael Levin
- Allen Discovery Center, Tufts University, Medford, MA 02155, USA; Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Yang J, Zhou S, Yang Z, Shi X, Liu H, Yang Z, Peng D, Ding Z, Ye S. Silencing of the T-type voltage-gated calcium channel α 1 subunit by fungus-mediated RNAi altered the structure of F-actin and caused defective behaviors in Ditylenchus destructor. Mol Biol Rep 2024; 51:673. [PMID: 38787479 DOI: 10.1007/s11033-024-09626-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND T-type calcium channels, characterized as low-voltage activated (LVA) calcium channels, play crucial physiological roles across a wide range of tissues, including both the neuronal and nonneuronal systems. Using in situ hybridization and RNA interference (RNAi) techniques in vitro, we previously identified the tissue distribution and physiological function of the T-type calcium channel α1 subunit (DdCα1G) in the plant-parasitic nematode Ditylenchus destructor. METHODS AND RESULTS To further characterize the functional role of DdCα1G, we employed a combination of immunohistochemistry and fungus-mediated RNAi and found that DdCα1G was clearly distributed in stylet-related tissue, oesophageal gland-related tissue, secretory-excretory duct-related tissue and male spicule-related tissue. Silencing DdCα1G led to impairments in the locomotion, feeding, reproductive ability and protein secretion of nematodes. To confirm the defects in behavior, we used phalloidin staining to examine muscle changes in DdCα1G-RNAi nematodes. Our observations demonstrated that defective behaviors are associated with related muscular atrophy. CONCLUSION Our findings provide a deeper understanding of the physiological functions of T-type calcium channels in plant-parasitic nematodes. The T-type calcium channel can be considered a promising target for sustainable nematode management practices.
Collapse
Affiliation(s)
- Jiahao Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Siyu Zhou
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Ziqi Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Xuqi Shi
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Haoran Liu
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
| | - Zhuhong Yang
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China
| | - Deliang Peng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zhong Ding
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China.
| | - Shan Ye
- College of Plant Protection, Hunan Agricultural University, Changsha, Hunan, 410128, China.
- Hunan Provincial Engineering & Technology Research Center for Biopesticide and Formulation Processing, Changsha, Hunan, 410128, China.
| |
Collapse
|
49
|
Xu F, Cai W, Liu B, Qiu Z, Zhang X. Natural L-type calcium channels antagonists from Chinese medicine. Chin Med 2024; 19:72. [PMID: 38773596 PMCID: PMC11107034 DOI: 10.1186/s13020-024-00944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
L-type calcium channels (LTCCs), the largest subfamily of voltage-gated calcium channels (VGCCs), are the main channels for Ca2+ influx during extracellular excitation. LTCCs are widely present in excitable cells, especially cardiac and cardiovascular smooth muscle cells, and participate in various Ca2+-dependent processes. LTCCs have been considered as worthy drug target for cardiovascular, neurological and psychological diseases for decades. Natural products from Traditional Chinese medicine (TCM) have shown the potential as new drugs for the treatment of LTCCs related diseases. In this review, the basic structure, function of LTCCs, and the related human diseases caused by structural or functional abnormalities of LTCCs, and the natural LTCCs antagonist and their potential usages were summarized.
Collapse
Affiliation(s)
- Fangfang Xu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Wanna Cai
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Bo Liu
- The Second Clinical College , Guangzhou University of Chinese Medicine, Guangzhou, 510006, People's Republic of China
| | - Zhenwen Qiu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| | - Xiaoqi Zhang
- Guangdong Provincial Engineering Research Center for Modernization of TCM, NMPA Key Laboratory for Quality Evaluation of TCM, Jinan University, Guangzhou, 510632, People's Republic of China.
| |
Collapse
|
50
|
Del Bianco D, Gentile R, Sallicandro L, Biagini A, Quellari PT, Gliozheni E, Sabbatini P, Ragonese F, Malvasi A, D’Amato A, Baldini GM, Trojano G, Tinelli A, Fioretti B. Electro-Metabolic Coupling of Cumulus-Oocyte Complex. Int J Mol Sci 2024; 25:5349. [PMID: 38791387 PMCID: PMC11120766 DOI: 10.3390/ijms25105349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Oocyte-cumulus cell interaction is essential for oocyte maturation and competence. The bidirectional crosstalk network mediated by gap junctions is fundamental for the metabolic cooperation between these cells. As cumulus cells exhibit a more glycolytic phenotype, they can provide metabolic substrates that the oocyte can use to produce ATP via oxidative phosphorylation. The impairment of mitochondrial activity plays a crucial role in ovarian aging and, thus, in fertility, determining the success or failure of assisted reproductive techniques. This review aims to deepen the knowledge about the electro-metabolic coupling of the cumulus-oocyte complex and to hypothesize a putative role of potassium channel modulators in order to improve fertility, promote intracellular Ca2+ influx, and increase the mitochondrial biogenesis and resulting ATP levels in cumulus cells.
Collapse
Affiliation(s)
- Diletta Del Bianco
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
| | - Rosaria Gentile
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| | - Luana Sallicandro
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Biagini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Paola Tiziana Quellari
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
- ASST Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy
| | - Elko Gliozheni
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Department of Medicine and Surgery, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Tirana, AL1005 Tirana, Albania
| | - Paola Sabbatini
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio Malvasi
- Department of Biomedical Sciences and Human Oncology, University of Bari, 70121 Bari, Italy;
| | - Antonio D’Amato
- 1st Unit of Obstetrics and Gynecology, University of Bari, 70121 Bari, Italy;
| | | | - Giuseppe Trojano
- Department of Maternal and Child Health, “Madonna delle Grazie” Hospital ASM, 75100 Matera, Italy;
| | - Andrea Tinelli
- Department of Obstetrics and Gynecology and CERICSAL (CEntro di RIcerca Clinico SALentino), Veris delli Ponti Hospital, Via Giuseppina delli Ponti, 73020 Scorrano, Lecce, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via dell’Elce di Sotto 8, 06132 Perugia, Italy; (D.D.B.); (R.G.); (L.S.); (A.B.); (P.T.Q.); (E.G.); (P.S.); (F.R.)
- Laboratorio Interdipartimentale di Fisiopatologia della Riproduzione, Università degli Studi di Perugia, Edificio C, Piano 3 P.zza Lucio Severi, 1, Sant’Andrea delle Fratte, 06132 Perugia, Italy
| |
Collapse
|