1
|
Shapiro IM, Risbud MV, Tang T, Landis WJ. Skeletal and dental tissue mineralization: The potential role of the endoplasmic reticulum/Golgi complex and the endolysosomal and autophagic transport systems. Bone 2025; 193:117390. [PMID: 39814250 DOI: 10.1016/j.bone.2025.117390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
This paper presents a review of the potential role of the endoplasmic reticulum/Golgi complex and intracellular vesicles in mediating events leading to or associated with vertebrate tissue mineralization. The possible importance of these organelles in this process is suggested by observations that calcium ions accumulate in the tubules and lacunae of the endoplasmic reticulum and Golgi. Similar levels of calcium ions (approaching millimolar) are present in vesicles derived from endosomes, lysosomes and autophagosomes. The cellular level of phosphate ions in these organelles is also high (millimolar). While the source of these ions for mineral formation has not been identified, there are sound reasons for considering that they may be liberated from mitochondria during the utilization of ATP for anabolic purposes, perhaps linked to matrix synthesis. Published studies indicate that calcium and phosphate ions or their clusters contained as cargo within the intracellular organelles noted above lead to formation of extracellular mineral. The mineral sequestered in mitochondria has been documented as an amorphous calcium phosphate. The ion-, ion cluster- or mineral-containing vesicles exit the cell in plasma membrane blebs, secretory lysosomes or possibly intraluminal vesicles. Such a cell-regulated process provides a means for the rapid transport of ions or mineral particles to the mineralization front of skeletal and dental tissues. Within the extracellular matrix, the ions or mineral may associate to form larger aggregates and potential mineral nuclei, and they may bind to collagen and other proteins. How cells of hard tissues perform their housekeeping and other biosynthetic functions while transporting the very large volumes of ions required for mineralization of the extracellular matrix is far from clear. Addressing this and related questions raised in this review suggests guidelines for further investigations of the intracellular processes promoting the mineralization of the skeletal and dental tissues.
Collapse
Affiliation(s)
- Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America.
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Tengteng Tang
- Center for Applied Biomechanics, Department of Mechanical and Aerospace Engineering, School of Engineering and Applied Science, University of Virginia, Charlottesville, VA, United States of America
| | - William J Landis
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California at San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
2
|
Jenkins PM, Bender KJ. Axon initial segment structure and function in health and disease. Physiol Rev 2025; 105:765-801. [PMID: 39480263 DOI: 10.1152/physrev.00030.2024] [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: 08/01/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
At the simplest level, neurons are structured to integrate synaptic input and perform computational transforms on that input, converting it into an action potential (AP) code. This process, converting synaptic input into AP output, typically occurs in a specialized region of the axon termed the axon initial segment (AIS). The AIS, as its name implies, is often contained to the first section of axon abutted to the soma and is home to a dizzying array of ion channels, attendant scaffolding proteins, intracellular organelles, extracellular proteins, and, in some cases, synapses. The AIS serves multiple roles as the final arbiter for determining if inputs are sufficient to evoke APs, as a gatekeeper that physically separates the somatodendritic domain from the axon proper, and as a regulator of overall neuronal excitability, dynamically tuning its size to best suit the needs of parent neurons. These complex roles have received considerable attention from experimentalists and theoreticians alike. Here, we review recent advances in our understanding of the AIS and its role in neuronal integration and polarity in health and disease.
Collapse
Affiliation(s)
- Paul M Jenkins
- Departments of Pharmacology and Psychiatry, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Kevin J Bender
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, California, United States
| |
Collapse
|
3
|
Ferns M, van der List D, Vierra NC, Lacey T, Murray K, Kirmiz M, Stewart RG, Sack JT, Trimmer JS. The Electrically Silent Kv5.1 Subunit Forms Heteromeric Kv2 Channels in Cortical Neurons and Confers Distinct Functional Properties. J Neurosci 2025; 45:e2293232025. [PMID: 39933932 PMCID: PMC11949482 DOI: 10.1523/jneurosci.2293-23.2025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Voltage-gated K+ channels of the Kv2 family are highly expressed in brain and play dual roles in regulating neuronal excitability and in organizing endoplasmic reticulum-plasma membrane (ER-PM) junctions. Studies in heterologous cells suggest that Kv2.1 and Kv2.2 co-assemble with "electrically silent" KvS subunits to form heterotetrameric channels with distinct biophysical properties, but the prevalence and localization of these channels in native neurons are unknown. Here, using mass spectrometry-based proteomics, we identified five KvS subunits as components of native Kv2.1 channels immunopurified from mouse brain of both sexes, the most abundant being Kv5.1. We found that Kv5.1 co-immunoprecipitates with Kv2.1 and to a lesser extent with Kv2.2 from brain lysates and that Kv5.1 protein levels are decreased by 70% in Kv2.1 knock-out mice and 95% in Kv2.1/Kv2.2 double knock-out mice. RNAscope and immunolabeling revealed that Kv5.1 is prominently expressed in neocortex, where it is detected in a substantial fraction of Kv2.1/Kv2.2 positive neurons in layers 2/3, 5, and 6. At the subcellular level, Kv5.1 protein is coclustered with Kv2.1 and Kv2.2 at presumptive ER-PM junctions on the soma and proximal dendrites of cortical neurons. Moreover, in addition to modifying channel conductance, we found that Kv2/Kv5.1 channels are less phosphorylated and insensitive to RY785, a potent and selective Kv2 channel inhibitor. Together, these findings demonstrate that KvS subunits create multiple Kv2 channel subtypes in brain. Most notably, Kv2/Kv5.1 channels are highly expressed in cortical neurons, where their unique properties likely modulate the critical conducting and nonconducting roles of Kv2 channels.
Collapse
Affiliation(s)
- Michael Ferns
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, California 95616
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Deborah van der List
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Nicholas C Vierra
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Taylor Lacey
- Department of Anesthesiology and Pain Medicine, University of California Davis, Davis, California 95616
| | - Karl Murray
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Michael Kirmiz
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Robert G Stewart
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| | - James S Trimmer
- Department of Physiology and Membrane Biology, University of California Davis, Davis, California 95616
| |
Collapse
|
4
|
Benedetti L, Fan R, Weigel AV, Moore AS, Houlihan PR, Kittisopikul M, Park G, Petruncio A, Hubbard PM, Pang S, Xu CS, Hess HF, Saalfeld S, Rangaraju V, Clapham DE, De Camilli P, Ryan TA, Lippincott-Schwartz J. Periodic ER-plasma membrane junctions support long-range Ca 2+ signal integration in dendrites. Cell 2025; 188:484-500.e22. [PMID: 39708809 DOI: 10.1016/j.cell.2024.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 11/01/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
Neuronal dendrites must relay synaptic inputs over long distances, but the mechanisms by which activity-evoked intracellular signals propagate over macroscopic distances remain unclear. Here, we discovered a system of periodically arranged endoplasmic reticulum-plasma membrane (ER-PM) junctions tiling the plasma membrane of dendrites at ∼1 μm intervals, interlinked by a meshwork of ER tubules patterned in a ladder-like array. Populated with Junctophilin-linked plasma membrane voltage-gated Ca2+ channels and ER Ca2+-release channels (ryanodine receptors), ER-PM junctions are hubs for ER-PM crosstalk, fine-tuning of Ca2+ homeostasis, and local activation of the Ca2+/calmodulin-dependent protein kinase II. Local spine stimulation activates the Ca2+ modulatory machinery, facilitating signal transmission and ryanodine-receptor-dependent Ca2+ release at ER-PM junctions over 20 μm away. Thus, interconnected ER-PM junctions support signal propagation and Ca2+ release from the spine-adjacent ER. The capacity of this subcellular architecture to modify both local and distant membrane-proximal biochemistry potentially contributes to dendritic computations.
Collapse
Affiliation(s)
| | - Ruolin Fan
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | | | | | | | | | - Grace Park
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | | | | | - Song Pang
- Yale School of Medicine, New Haven, CT 06510, USA
| | - C Shan Xu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Harald F Hess
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | | | - Vidhya Rangaraju
- Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | | | - Pietro De Camilli
- Department of Neuroscience, Department of Cell Biology, Program in Cellular Neuroscience Neurodegeneration and Repair, Howard Hughes Medical Institute, New Haven, CT 06510, USA
| | - Timothy A Ryan
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA; Weill Cornell Medicine, Department of Biochemistry, New York, NY 10065, USA.
| | | |
Collapse
|
5
|
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
|
6
|
Johnson B, Iuliano M, Lam TT, Biederer T, De Camilli PV. A complex of the lipid transport ER proteins TMEM24 and C2CD2 with band 4.1 at cell-cell contacts. J Cell Biol 2024; 223:e202311137. [PMID: 39158698 PMCID: PMC11334333 DOI: 10.1083/jcb.202311137] [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: 11/21/2023] [Revised: 04/23/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Junctions between the ER and plasma membrane (PM) are implicated in calcium homeostasis, non-vesicular lipid transfer, and other cellular functions. Two ER proteins that function both as tethers to the PM via a polybasic C-terminus motif and as phospholipid transporters are brain-enriched TMEM24 (C2CD2L) and its paralog C2CD2. We report that both proteins also form a complex with band 4.1 family members, which in turn bind PM proteins including cell adhesion molecules such as SynCAM 1. This complex enriches TMEM24 and C2CD2 containing ER/PM junctions at sites of cell contacts. Dynamic properties of TMEM24-dependent ER/PM junctions are impacted when band 4.1 is part of the junction, as TMEM24 at cell-adjacent ER/PM junctions is not shed from the PM by calcium rise, unlike TMEM24 at non-cell adjacent junctions. Lipid transport between the ER and the PM by TMEM24 and C2CD2 at sites where cells, including neurons, contact other cells may participate in adaptive responses to cell contact-dependent signaling.
Collapse
Affiliation(s)
- Ben Johnson
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Maria Iuliano
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA
- Department of Keck MS and Proteomics Resource, Yale University School of Medicine, New Haven, CT, USA
| | - Thomas Biederer
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Pietro V De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
Vierra NC. Compartmentalized signaling in the soma: Coordination of electrical and protein kinase A signaling at neuronal ER-plasma membrane junctions. Bioessays 2024; 46:e2400126. [PMID: 39268818 DOI: 10.1002/bies.202400126] [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: 05/28/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024]
Abstract
Neuronal information processing depends on converting membrane depolarizations into compartmentalized biochemical signals that can modify neuronal activity and structure. However, our understanding of how neurons translate electrical signals into specific biochemical responses remains limited, especially in the soma where gene expression and ion channel function are crucial for neuronal activity. Here, I emphasize the importance of physically compartmentalizing action potential-triggered biochemical reactions within the soma. Emerging evidence suggests that somatic endoplasmic reticulum-plasma membrane (ER-PM) junctions are specialized organelles that coordinate electrical and biochemical signaling. The juxtaposition of ion channels and signaling proteins at a prominent subset of these sites enables compartmentalized calcium and cAMP-dependent protein kinase (PKA) signaling. I explore the hypothesis that these PKA-containing ER-PM junctions serve as critical sites for translating membrane depolarizations into PKA signals and identify key gaps in knowledge of the assembly, regulation, and neurobiological functions of this somatic signaling system.
Collapse
Affiliation(s)
- Nicholas C Vierra
- School of Biological Sciences, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
8
|
Maciąg F, Chhikara A, Heine M. Calcium channel signalling at neuronal endoplasmic reticulum-plasma membrane junctions. Biochem Soc Trans 2024; 52:1617-1629. [PMID: 38934485 PMCID: PMC11668288 DOI: 10.1042/bst20230819] [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: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024]
Abstract
Neurons are highly specialised cells that need to relay information over long distances and integrate signals from thousands of synaptic inputs. The complexity of neuronal function is evident in the morphology of their plasma membrane (PM), by far the most intricate of all cell types. Yet, within the neuron lies an organelle whose architecture adds another level to this morphological sophistication - the endoplasmic reticulum (ER). Neuronal ER is abundant in the cell body and extends to distant axonal terminals and postsynaptic dendritic spines. It also adopts specialised structures like the spine apparatus in the postsynapse and the cisternal organelle in the axon initial segment. At membrane contact sites (MCSs) between the ER and the PM, the two membranes come in close proximity to create hubs of lipid exchange and Ca2+ signalling called ER-PM junctions. The development of electron and light microscopy techniques extended our knowledge on the physiological relevance of ER-PM MCSs. Equally important was the identification of ER and PM partners that interact in these junctions, most notably the STIM-ORAI and VAP-Kv2.1 pairs. The physiological functions of ER-PM junctions in neurons are being increasingly explored, but their molecular composition and the role in the dynamics of Ca2+ signalling are less clear. This review aims to outline the current state of research on the topic of neuronal ER-PM contacts. Specifically, we will summarise the involvement of different classes of Ca2+ channels in these junctions, discuss their role in neuronal development and neuropathology and propose directions for further research.
Collapse
Affiliation(s)
- Filip Maciąg
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Arun Chhikara
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| | - Martin Heine
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Hanns-Dieter Hüsch Weg 15, 55128 Mainz, Germany
| |
Collapse
|
9
|
Smith CC, Nascimento F, Özyurt MG, Beato M, Brownstone RM. Kv2 channels do not function as canonical delayed rectifiers in spinal motoneurons. iScience 2024; 27:110444. [PMID: 39148717 PMCID: PMC11325356 DOI: 10.1016/j.isci.2024.110444] [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: 02/09/2024] [Revised: 04/29/2024] [Accepted: 07/01/2024] [Indexed: 08/17/2024] Open
Abstract
The increased muscular force output required for some behaviors is achieved via amplification of motoneuron output via cholinergic C-bouton synapses. Work in neonatal mouse motoneurons suggested that modulation of currents mediated by post-synaptically clustered KV2.1 channels is crucial to C-bouton amplification. By focusing on more mature motoneurons, we show that conditional knockout of KV2.1 channels minimally affects either excitability or response to exogenously applied muscarine. Similarly, unlike in neonatal motoneurons or cortical pyramidal neurons, pharmacological blockade of KV2 currents has minimal effect on mature motoneuron firing in vitro. Furthermore, in vivo amplification of electromyography activity and high-force task performance was unchanged following KV2.1 knockout. Finally, we show that KV2.2 is also expressed by spinal motoneurons, colocalizing with KV2.1 opposite C-boutons. We suggest that the primary function of KV2 proteins in motoneurons is non-conducting and that KV2.2 can function in this role in the absence of KV2.1.
Collapse
Affiliation(s)
- Calvin C. Smith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Filipe Nascimento
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - M. Görkem Özyurt
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Marco Beato
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London WC1E 6BT, UK
| | - Robert M. Brownstone
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
10
|
Sun S, Zhao G, Jia M, Jiang Q, Li S, Wang H, Li W, Wang Y, Bian X, Zhao YG, Huang X, Yang G, Cai H, Pastor-Pareja JC, Ge L, Zhang C, Hu J. Stay in touch with the endoplasmic reticulum. SCIENCE CHINA. LIFE SCIENCES 2024; 67:230-257. [PMID: 38212460 DOI: 10.1007/s11427-023-2443-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/28/2023] [Indexed: 01/13/2024]
Abstract
The endoplasmic reticulum (ER), which is composed of a continuous network of tubules and sheets, forms the most widely distributed membrane system in eukaryotic cells. As a result, it engages a variety of organelles by establishing membrane contact sites (MCSs). These contacts regulate organelle positioning and remodeling, including fusion and fission, facilitate precise lipid exchange, and couple vital signaling events. Here, we systematically review recent advances and converging themes on ER-involved organellar contact. The molecular basis, cellular influence, and potential physiological functions for ER/nuclear envelope contacts with mitochondria, Golgi, endosomes, lysosomes, lipid droplets, autophagosomes, and plasma membrane are summarized.
Collapse
Affiliation(s)
- Sha Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gan Zhao
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Mingkang Jia
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haibin Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenjing Li
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunyun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xin Bian
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yan G Zhao
- Brain Research Center, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ge Yang
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jose C Pastor-Pareja
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Neurosciences, Consejo Superior de Investigaciones Cientfflcas-Universidad Miguel Hernandez, San Juan de Alicante, 03550, Spain.
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
11
|
Leek AN, Quinn JA, Krapf D, Tamkun MM. GLT-1a glutamate transporter nanocluster localization is associated with astrocytic actin and neuronal Kv2 clusters at sites of neuron-astrocyte contact. Front Cell Dev Biol 2024; 12:1334861. [PMID: 38362041 PMCID: PMC10867268 DOI: 10.3389/fcell.2024.1334861] [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: 11/07/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction: Astrocytic GLT-1 glutamate transporters ensure the fidelity of glutamic neurotransmission by spatially and temporally limiting glutamate signals. The ability to limit neuronal hyperactivity relies on the localization and diffusion of GLT-1 on the astrocytic surface, however, little is known about the underlying mechanisms. We show that two isoforms of GLT-1, GLT-1a and GLT-1b, form nanoclusters on the surface of transfected astrocytes and HEK-293 cells. Methods: We used both fixed and live cell super-resolution imaging of fluorescent protein and epitope tagged proteins in co-cultures of rat astrocytes and neurons. Immunofluorescence techniques were also used. GLT1 diffusion was assessed via single particle tracking and fluorescence recovery after photobleach (FRAP). Results: We found GLT-1a, but not GLT-1b, nanoclusters concentrated adjacent to actin filaments which was maintained after addition of glutamate. GLT-1a nanocluster concentration near actin filaments was prevented by expression of a cytosolic GLT-1a C-terminus, suggesting the C-terminus is involved in the localization adjacent to cortical actin. Using super-resolution imaging, we show that astrocytic GLT-1a and actin co-localize in net-like structures around neuronal Kv2.1 clusters at points of neuron/astrocyte contact. Conclusion: Overall, these data describe a novel relationship between GLT-1a and cortical actin filaments, which localizes GLT-1a near neuronal structures responsive to ischemic insult.
Collapse
Affiliation(s)
- Ashley N. Leek
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States
| | - Josiah A. Quinn
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO, United States
| | - Michael M. Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
12
|
Ferns M, van der List D, Vierra NC, Lacey T, Murray K, Kirmiz M, Stewart RG, Sack JT, Trimmer JS. Electrically silent KvS subunits associate with native Kv2 channels in brain and impact diverse properties of channel function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577135. [PMID: 38328147 PMCID: PMC10849721 DOI: 10.1101/2024.01.25.577135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Voltage-gated K+ channels of the Kv2 family are highly expressed in brain and play dual roles in regulating neuronal excitability and in organizing endoplasmic reticulum - plasma membrane (ER-PM) junctions. Studies in heterologous cells suggest that the two pore-forming alpha subunits Kv2.1 and Kv2.2 assemble with "electrically silent" KvS subunits to form heterotetrameric channels with distinct biophysical properties. Here, using mass spectrometry-based proteomics, we identified five KvS subunits as components of native Kv2.1 channels immunopurified from mouse brain, the most abundant being Kv5.1. We found that Kv5.1 co-immunoprecipitates with Kv2.1 and to a lesser extent with Kv2.2 from brain lysates, and that Kv5.1 protein levels are decreased by 70% in Kv2.1 knockout mice and 95% in Kv2.1/2.2 double knockout mice. Multiplex immunofluorescent labelling of rodent brain sections revealed that in neocortex Kv5.1 immunolabeling is apparent in a large percentage of Kv2.1 and Kv2.2-positive layer 2/3 neurons, and in a smaller percentage of layer 5 and 6 neurons. At the subcellular level, Kv5.1 is co-clustered with Kv2.1 and Kv2.2 at ER-PM junctions in cortical neurons, although clustering of Kv5.1-containing channels is reduced relative to homomeric Kv2 channels. We also found that in heterologous cells coexpression with Kv5.1 reduces the clustering and alters the pharmacological properties of Kv2.1 channels. Together, these findings demonstrate that the Kv5.1 electrically silent subunit is a component of a substantial fraction of native brain Kv2 channels, and that its incorporation into heteromeric channels can impact diverse aspects of Kv2 channel function.
Collapse
Affiliation(s)
- Michael Ferns
- Dept. of Anesthesiology and Pain Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Deborah van der List
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Nicholas C. Vierra
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Taylor Lacey
- Dept. of Anesthesiology and Pain Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Karl Murray
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
- Dept. of Psychiatry and Behavioral Sciences, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Michael Kirmiz
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Robert G. Stewart
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Jon T. Sack
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - James S. Trimmer
- Dept. of Physiology and Membrane Biology, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
13
|
Johnson B, Iuliano M, Lam T, Biederer T, De Camilli P. A complex of the lipid transport ER proteins TMEM24 and C2CD2 with band 4.1 at cell-cell contacts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570396. [PMID: 38106008 PMCID: PMC10723409 DOI: 10.1101/2023.12.06.570396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Junctions between the ER and the plasma membrane (ER/PM junctions) are implicated in calcium homeostasis, non-vesicular lipid transfer and other cellular functions. Two ER proteins that function both as membrane tethers to the PM via a polybasic motif in their C-terminus and as phospholipid transporters are brain-enriched TMEM24 (C2CD2L) and its paralog C2CD2. Based on an unbiased proximity ligation analysis, we found that both proteins can also form a complex with band 4.1 family members, which in turn can bind a variety of plasma membrane proteins including cell adhesion molecules such as SynCAM 1. This complex results in the enrichment of TMEM24 and C2CD2 containing ER/PM junctions at sites of cell contacts. Dynamic properties of TMEM24-dependent ER/PM contacts are impacted when in complex as TMEM24 present at cell adjacent junctions is not shed by calcium rise, unlike TMEM24 at non-cell adjacent junctions. These findings suggest that cell-contact interactions control ER/PM junctions via TMEM24 complexes involving band 4.1 proteins.
Collapse
Affiliation(s)
- Ben Johnson
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Maria Iuliano
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111
| | - TuKiet Lam
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Keck MS and Proteomics Resource, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Thomas Biederer
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
14
|
Dixon RE, Trimmer JS. Endoplasmic Reticulum-Plasma Membrane Junctions as Sites of Depolarization-Induced Ca 2+ Signaling in Excitable Cells. Annu Rev Physiol 2023; 85:217-243. [PMID: 36202100 PMCID: PMC9918718 DOI: 10.1146/annurev-physiol-032122-104610] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Membrane contact sites between endoplasmic reticulum (ER) and plasma membrane (PM), or ER-PM junctions, are found in all eukaryotic cells. In excitable cells they play unique roles in organizing diverse forms of Ca2+ signaling as triggered by membrane depolarization. ER-PM junctions underlie crucial physiological processes such as excitation-contraction coupling, smooth muscle contraction and relaxation, and various forms of activity-dependent signaling and plasticity in neurons. In many cases the structure and molecular composition of ER-PM junctions in excitable cells comprise important regulatory feedback loops linking depolarization-induced Ca2+ signaling at these sites to the regulation of membrane potential. Here, we describe recent findings on physiological roles and molecular composition of native ER-PM junctions in excitable cells. We focus on recent studies that provide new insights into canonical forms of depolarization-induced Ca2+ signaling occurring at junctional triads and dyads of striated muscle, as well as the diversity of ER-PM junctions in these cells and in smooth muscle and neurons.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California, USA;
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
15
|
Vullhorst D, Bloom MS, Akella N, Buonanno A. ER-PM Junctions on GABAergic Interneurons Are Organized by Neuregulin 2/VAP Interactions and Regulated by NMDA Receptors. Int J Mol Sci 2023; 24:2908. [PMID: 36769244 PMCID: PMC9917868 DOI: 10.3390/ijms24032908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Neuregulins (NRGs) signal via ErbB receptors to regulate neural development, excitability, synaptic and network activity, and behaviors relevant to psychiatric disorders. Bidirectional signaling between NRG2/ErbB4 and NMDA receptors is thought to homeostatically regulate GABAergic interneurons in response to increased excitatory neurotransmission or elevated extracellular glutamate levels. Unprocessed proNRG2 forms discrete clusters on cell bodies and proximal dendrites that colocalize with the potassium channel Kv2.1 at specialized endoplasmic reticulum-plasma membrane (ER-PM) junctions, and NMDA receptor activation triggers rapid dissociation from ER-PM junctions and ectodomain shedding by ADAM10. Here, we elucidate the mechanistic basis of proNRG2 clustering at ER-PM junctions and its regulation by NMDA receptors. Importantly, we demonstrate that proNRG2 promotes the formation of ER-PM junctions by directly binding the ER-resident membrane tether VAP, like Kv2.1. The proNRG2 intracellular domain harbors two non-canonical, low-affinity sites that cooperatively mediate VAP binding. One of these is a cryptic and phosphorylation-dependent VAP binding motif that is dephosphorylated following NMDA receptor activation, thus revealing how excitatory neurotransmission promotes the dissociation of proNRG2 from ER-PM junctions. Therefore, proNRG2 and Kv2.1 can independently function as VAP-dependent organizers of neuronal ER-PM junctions. Based on these and prior studies, we propose that proNRG2 and Kv2.1 serve as co-regulated downstream effectors of NMDA receptors to homeostatically regulate GABAergic interneurons.
Collapse
Affiliation(s)
- Detlef Vullhorst
- Section on Molecular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
16
|
Chen Z, Hu W, Mendez MJ, Gossman ZC, Chomyk A, Boylan BT, Kidd GJ, Phares TW, Bergmann CC, Trapp BD. Neuroprotection by Preconditioning in Mice is Dependent on MyD88-Mediated CXCL10 Expression in Endothelial Cells. ASN Neuro 2023; 15:17590914221146365. [PMID: 36591943 PMCID: PMC9810995 DOI: 10.1177/17590914221146365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 01/03/2023] Open
Abstract
The central nervous system (CNS) can be preconditioned to resist damage by peripheral pretreatment with low-dose gram-negative bacterial endotoxin lipopolysaccharide (LPS). Underlying mechanisms associated with transient protection of the cerebral cortex against traumatic brain injury include increased neuronal production of antiapoptotic and neurotrophic molecules, microglial-mediated displacement of inhibitory presynaptic terminals innervating the soma of cortical projection neurons, and synchronized firing of cortical projection neurons. However, the cell types and signaling responsible for these neuronal and microglial changes are unknown. A fundamental question is whether LPS penetrates the CNS or acts on the luminal surface of brain endothelial cells, thereby triggering an indirect parenchymal neuroprotective response. The present study shows that a low-dose intraperitoneal LPS treatment increases brain endothelial cell activation markers CD54, but does not open the blood-brain barrier or alter brain endothelial cell tight junctions as assessed by electron microscopy. NanoString nCounter transcript analyses of CD31-positive brain endothelial cells further revealed significant upregulation of Cxcl10, C3, Ccl2, Il1β, Cxcl2, and Cxcl1, consistent with identification of myeloid differentiation primary response 88 (MyD88) as a regulator of these transcripts by pathway analysis. Conditional genetic endothelial cell gene ablation approaches demonstrated that both MyD88-dependent Toll-like receptor 4 (TLR4) signaling and Cxcl10 expression are essential for LPS-induced neuroprotection and microglial activation. These results suggest that C-X-C motif chemokine ligand 10 (CXCL10) production by endothelial cells in response to circulating TLR ligands may directly or indirectly signal to CXCR3 on neurons and/or microglia. Targeted activation of brain endothelial receptors may thus provide an attractive approach for inducing transient neuroprotection.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Weiwei Hu
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
- Department of Pharmacology, School of Basic Medical Sciences,
Zhejiang
University, Hangzhou, China
| | - Mynor J. Mendez
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Zachary C. Gossman
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Anthony Chomyk
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Brendan T. Boylan
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
- Department of Pathology, Case Western Reserve University School of
Medicine, Cleveland, OH, USA
| | - Grahame J. Kidd
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Timothy W. Phares
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Cornelia C. Bergmann
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| | - Bruce D. Trapp
- Department of Neurosciences, Lerner Research Institute,
Cleveland
Clinic, Cleveland, OH, USA
| |
Collapse
|
17
|
Forzisi E, Sesti F. Non-conducting functions of ion channels: The case of integrin-ion channel complexes. Channels (Austin) 2022; 16:185-197. [PMID: 35942524 PMCID: PMC9364710 DOI: 10.1080/19336950.2022.2108565] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Started as an academic curiosity more than two decades ago, the idea that ion channels can regulate cellular processes in ways that do not depend on their conducting properties (non-ionic functions) gained traction and is now a flourishing area of research. Channels can regulate physiological processes including actin cytoskeletal remodeling, cell motility, excitation-contraction coupling, non-associative learning and embryogenesis, just to mention some, through non-ionic functions. When defective, non-ionic functions can give rise to channelopathies involved in cancer, neurodegenerative disease and brain trauma. Ion channels exert their non-ionic functions through a variety of mechanisms that range from physical coupling with other proteins, to possessing enzymatic activity, to assembling with signaling molecules. In this article, we take stock of the field and review recent findings. The concept that emerges, is that one of the most common ways through which channels acquire non-ionic attributes, is by assembling with integrins. These integrin-channel complexes exhibit broad genotypic and phenotypic heterogeneity and reveal a pleiotropic nature, as they appear to be capable of influencing both physiological and pathological processes.
Collapse
Affiliation(s)
- Elena Forzisi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| |
Collapse
|
18
|
Piccialli I, Sisalli MJ, de Rosa V, Boscia F, Tedeschi V, Secondo A, Pannaccione A. Increased K V2.1 Channel Clustering Underlies the Reduction of Delayed Rectifier K + Currents in Hippocampal Neurons of the Tg2576 Alzheimer's Disease Mouse. Cells 2022; 11:cells11182820. [PMID: 36139395 PMCID: PMC9497218 DOI: 10.3390/cells11182820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the progressive deterioration of cognitive functions. Cortical and hippocampal hyperexcitability intervenes in the pathological derangement of brain activity leading to cognitive decline. As key regulators of neuronal excitability, the voltage-gated K+ channels (KV) might play a crucial role in the AD pathophysiology. Among them, the KV2.1 channel, the main α subunit mediating the delayed rectifier K+ currents (IDR) and controlling the intrinsic excitability of pyramidal neurons, has been poorly examined in AD. In the present study, we investigated the KV2.1 protein expression and activity in hippocampal neurons from the Tg2576 mouse, a widely used transgenic model of AD. To this aim we performed whole-cell patch-clamp recordings, Western blotting, and immunofluorescence analyses. Our Western blotting results reveal that KV2.1 was overexpressed in the hippocampus of 3-month-old Tg2576 mice and in primary hippocampal neurons from Tg2576 mouse embryos compared with the WT counterparts. Electrophysiological experiments unveiled that the whole IDR were reduced in the Tg2576 primary neurons compared with the WT neurons, and that this reduction was due to the loss of the KV2.1 current component. Moreover, we found that the reduction of the KV2.1-mediated currents was due to increased channel clustering, and that glutamate, a stimulus inducing KV2.1 declustering, was able to restore the IDR to levels comparable to those of the WT neurons. These findings add new information about the dysregulation of ionic homeostasis in the Tg2576 AD mouse model and identify KV2.1 as a possible player in the AD-related alterations of neuronal excitability.
Collapse
|
19
|
Activity-dependent endoplasmic reticulum Ca 2+ uptake depends on Kv2.1-mediated endoplasmic reticulum/plasma membrane junctions to promote synaptic transmission. Proc Natl Acad Sci U S A 2022; 119:e2117135119. [PMID: 35862456 PMCID: PMC9335237 DOI: 10.1073/pnas.2117135119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The endoplasmic reticulum (ER) extends throughout the neuron as a continuous organelle, and its dysfunction is associated with several neurological disorders. During electrical activity, the ER takes up Ca2+ from the cytosol, which has been shown to support synaptic transmission. This close choreography of ER Ca2+ uptake with electrical activity suggests functional coupling of the ER to sources of voltage-gated Ca2+ entry through an unknown mechanism. We report that a nonconducting role for Kv2.1 through its ER binding domain is necessary for ER Ca2+ uptake during neuronal activity. Loss of Kv2.1 profoundly disables neurotransmitter release without altering presynaptic voltage. This suggests that Kv2.1-mediated signaling hubs play an important neurobiological role in Ca2+ handling and synaptic transmission independent of ion conduction. The endoplasmic reticulum (ER) forms a continuous and dynamic network throughout a neuron, extending from dendrites to axon terminals, and axonal ER dysfunction is implicated in several neurological disorders. In addition, tight junctions between the ER and plasma membrane (PM) are formed by several molecules including Kv2 channels, but the cellular functions of many ER-PM junctions remain unknown. Recently, dynamic Ca2+ uptake into the ER during electrical activity was shown to play an essential role in synaptic transmission. Our experiments demonstrate that Kv2.1 channels are necessary for enabling ER Ca2+ uptake during electrical activity, as knockdown (KD) of Kv2.1 rendered both the somatic and axonal ER unable to accumulate Ca2+ during electrical stimulation. Moreover, our experiments demonstrate that the loss of Kv2.1 in the axon impairs synaptic vesicle fusion during stimulation via a mechanism unrelated to voltage. Thus, our data demonstrate that a nonconducting role of Kv2.1 exists through its binding to the ER protein VAMP-associated protein (VAP), which couples ER Ca2+ uptake with electrical activity. Our results further suggest that Kv2.1 has a critical function in neuronal cell biology for Ca2+ handling independent of voltage and reveals a critical pathway for maintaining ER lumen Ca2+ levels and efficient neurotransmitter release. Taken together, these findings reveal an essential nonclassical role for both Kv2.1 and the ER-PM junctions in synaptic transmission.
Collapse
|
20
|
Khan S. Endoplasmic Reticulum in Metaplasticity: From Information Processing to Synaptic Proteostasis. Mol Neurobiol 2022; 59:5630-5655. [PMID: 35739409 DOI: 10.1007/s12035-022-02916-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/05/2022] [Indexed: 11/29/2022]
Abstract
The ER (endoplasmic reticulum) is a Ca2+ reservoir and the unique protein-synthesizing machinery which is distributed throughout the neuron and composed of multiple different structural domains. One such domain is called EMC (endoplasmic reticulum membrane protein complex), pleiotropic nature in cellular functions. The ER/EMC position inside the neurons unmasks its contribution to synaptic plasticity via regulating various cellular processes from protein synthesis to Ca2+ signaling. Since presynaptic Ca2+ channels and postsynaptic ionotropic receptors are organized into the nanodomains, thus ER can be a crucial player in establishing TMNCs (transsynaptic molecular nanocolumns) to shape efficient neural communications. This review hypothesized that ER is not only involved in stress-mediated neurodegeneration but also axon regrowth, remyelination, neurotransmitter switching, information processing, and regulation of pre- and post-synaptic functions. Thus ER might not only be a protein-synthesizing and quality control machinery but also orchestrates plasticity of plasticity (metaplasticity) within the neuron to execute higher-order brain functions and neural repair.
Collapse
Affiliation(s)
- Shumsuzzaman Khan
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
21
|
Yang HQ, Echeverry FA, ElSheikh A, Gando I, Anez Arredondo S, Samper N, Cardozo T, Delmar M, Shyng SL, Coetzee WA. Subcellular trafficking and endocytic recycling of K ATP channels. Am J Physiol Cell Physiol 2022; 322:C1230-C1247. [PMID: 35508187 PMCID: PMC9169827 DOI: 10.1152/ajpcell.00099.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 11/22/2022]
Abstract
Sarcolemmal/plasmalemmal ATP-sensitive K+ (KATP) channels have key roles in many cell types and tissues. Hundreds of studies have described how the KATP channel activity and ATP sensitivity can be regulated by changes in the cellular metabolic state, by receptor signaling pathways and by pharmacological interventions. These alterations in channel activity directly translate to alterations in cell or tissue function, that can range from modulating secretory responses, such as insulin release from pancreatic β-cells or neurotransmitters from neurons, to modulating contractile behavior of smooth muscle or cardiac cells to elicit alterations in blood flow or cardiac contractility. It is increasingly becoming apparent, however, that KATP channels are regulated beyond changes in their activity. Recent studies have highlighted that KATP channel surface expression is a tightly regulated process with similar implications in health and disease. The surface expression of KATP channels is finely balanced by several trafficking steps including synthesis, assembly, anterograde trafficking, membrane anchoring, endocytosis, endocytic recycling, and degradation. This review aims to summarize the physiological and pathophysiological implications of KATP channel trafficking and mechanisms that regulate KATP channel trafficking. A better understanding of this topic has potential to identify new approaches to develop therapeutically useful drugs to treat KATP channel-related diseases.
Collapse
Affiliation(s)
- Hua-Qian Yang
- Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China
| | | | - Assmaa ElSheikh
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
- Department of Medical Biochemistry, Tanta University, Tanta, Egypt
| | - Ivan Gando
- Department of Pathology, NYU School of Medicine, New York, New York
| | | | - Natalie Samper
- Department of Pathology, NYU School of Medicine, New York, New York
| | - Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - Mario Delmar
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
- Department of Medicine, NYU School of Medicine, New York, New York
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| | - William A Coetzee
- Department of Pathology, NYU School of Medicine, New York, New York
- Department of Neuroscience & Physiology, NYU School of Medicine, New York, New York
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
22
|
Xu Z, Khan S, Schnicker NJ, Baker S. Pentameric assembly of the Kv2.1 tetramerization domain. Acta Crystallogr D Struct Biol 2022; 78:792-802. [PMID: 35647925 PMCID: PMC9159280 DOI: 10.1107/s205979832200568x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/25/2022] [Indexed: 11/25/2022] Open
Abstract
The Kv family of voltage-gated potassium channels regulate neuronal excitability. The biophysical characteristics of Kv channels can be matched to the needs of different neurons by forming homotetrameric or heterotetrameric channels within one of four subfamilies. The cytoplasmic tetramerization (T1) domain plays a major role in dictating the compatibility of different Kv subunits. The only Kv subfamily lacking a representative structure of the T1 domain is the Kv2 family. Here, X-ray crystallography was used to solve the structure of the human Kv2.1 T1 domain. The structure is similar to those of other T1 domains, but surprisingly formed a pentamer instead of a tetramer. In solution the Kv2.1 T1 domain also formed a pentamer, as determined by inline SEC-MALS-SAXS and negative-stain electron microscopy. The Kv2.1 T1-T1 interface involves electrostatic interactions, including a salt bridge formed by the negative charges in a previously described CDD motif, and inter-subunit coordination of zinc. It is shown that zinc binding is important for stability. In conclusion, the Kv2.1 T1 domain behaves differently from the other Kv T1 domains, which may reflect the versatility of Kv2.1, which can assemble with the regulatory KvS subunits and scaffold ER-plasma membrane contacts.
Collapse
Affiliation(s)
- Zhen Xu
- Protein and Crystallography Facility, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | - Saif Khan
- Protein and Crystallography Facility, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | - Nicholas J. Schnicker
- Protein and Crystallography Facility, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | - Sheila Baker
- Department of Biochemistry and Molecular Biology, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| |
Collapse
|
23
|
Maverick EE, Tamkun MM. High spatial density is associated with non-conducting Kv channels from two families. Biophys J 2022; 121:755-768. [PMID: 35101417 PMCID: PMC8943702 DOI: 10.1016/j.bpj.2022.01.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/14/2021] [Accepted: 01/25/2022] [Indexed: 11/02/2022] Open
Abstract
Ion channels are well known for their ability to regulate the cell membrane potential. However, many ion channels also have functions that do not involve ion conductance. Kv2 channels are one family of ion channels whose non-conducting functions are central to mammalian cell physiology. Kv2.1 and Kv2.2 channels form stable contact sites between the endoplasmic reticulum and plasma membrane via an interaction with endoplasmic reticulum resident proteins. To perform this structural role, Kv2 channels are expressed at extremely high densities on the plasma membranes of many cell types, including central pyramidal neurons, α-motoneurons, and smooth muscle cells. Research from our lab and others has shown that the majority of these plasma membrane Kv2.1 channels do not conduct potassium in response to depolarization. The mechanism of this channel silencing is unknown but is thought to be dependent on channel density in the membrane. Furthermore, the prevalence of a non-conducting population of Kv2.2 channels has not been directly tested. In this work we make improved measurements of the numbers of conducting and non-conducting Kv2.1 channels expressed in HEK293 cells and expand the investigation of non-conducting channels to three additional Kv α-subunits: Kv2.2, Kv1.4, and Kv1.5. By comparing the numbers of gating and conducting channels in individual HEK293 cells, we found that on average, only 50% of both Kv2.1 and Kv2.2 channels conducted potassium and, as previously suggested, that fraction decreased with increased channel density in the plasma membrane. At the highest spatial densities tested, which are comparable with those found at Kv2 clusters in situ, only 20% of Kv2.1 and Kv2.2 channels conducted potassium. We also show for the first time that Kv1.4 and Kv1.5 exhibit density-dependent silencing, suggesting that this phenomenon has an underlying mechanism that is shared by Kv channels from multiple families.
Collapse
Affiliation(s)
- Emily E. Maverick
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado,Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado
| | - Michael M. Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado,Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, Colorado,Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado,Corresponding author
| |
Collapse
|
24
|
Dickson EJ. Phosphoinositide transport and metabolism at membrane contact sites. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159107. [PMID: 34995791 PMCID: PMC9662651 DOI: 10.1016/j.bbalip.2021.159107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/18/2022]
Abstract
Phosphoinositides are a family of signaling lipids that play a profound role in regulating protein function at the membrane-cytosol interface of all cellular membranes. Underscoring their importance, mutations or alterations in phosphoinositide metabolizing enzymes lead to host of developmental, neurodegenerative, and metabolic disorders that are devastating for human health. In addition to lipid enzymes, phosphoinositide metabolism is regulated and controlled at membrane contact sites (MCS). Regions of close opposition typically between the ER and other cellular membranes, MCS are non-vesicular lipid transport portals that engage in extensive communication to influence organelle homeostasis. This review focuses on lipid transport, specifically phosphoinositide lipid transport and metabolism at MCS.
Collapse
Affiliation(s)
- Eamonn J Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, United States of America.
| |
Collapse
|
25
|
Ion Channel Partnerships: Odd and Not-So-Odd Couples Controlling Neuronal Ion Channel Function. Int J Mol Sci 2022; 23:ijms23041953. [PMID: 35216068 PMCID: PMC8878034 DOI: 10.3390/ijms23041953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
The concerted function of the large number of ion channels expressed in excitable cells, including brain neurons, shapes diverse signaling events by controlling the electrical properties of membranes. It has long been recognized that specific groups of ion channels are functionally coupled in mediating ionic fluxes that impact membrane potential, and that these changes in membrane potential impact ion channel gating. Recent studies have identified distinct sets of ion channels that can also physically and functionally associate to regulate the function of either ion channel partner beyond that afforded by changes in membrane potential alone. Here, we review canonical examples of such ion channel partnerships, in which a Ca2+ channel is partnered with a Ca2+-activated K+ channel to provide a dedicated route for efficient coupling of Ca2+ influx to K+ channel activation. We also highlight examples of non-canonical ion channel partnerships between Ca2+ channels and voltage-gated K+ channels that are not intrinsically Ca2+ sensitive, but whose partnership nonetheless yields enhanced regulation of one or the other ion channel partner. We also discuss how these ion channel partnerships can be shaped by the subcellular compartments in which they are found and provide perspectives on how recent advances in techniques to identify proteins in close proximity to one another in native cells may lead to an expanded knowledge of other ion channel partnerships.
Collapse
|
26
|
Sahu G, Turner RW. The Molecular Basis for the Calcium-Dependent Slow Afterhyperpolarization in CA1 Hippocampal Pyramidal Neurons. Front Physiol 2022; 12:759707. [PMID: 35002757 PMCID: PMC8730529 DOI: 10.3389/fphys.2021.759707] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 12/02/2022] Open
Abstract
Neuronal signal transmission depends on the frequency, pattern, and timing of spike output, each of which are shaped by spike afterhyperpolarizations (AHPs). There are classically three post-spike AHPs of increasing duration categorized as fast, medium and slow AHPs that hyperpolarize a cell over a range of 10 ms to 30 s. Intensive early work on CA1 hippocampal pyramidal cells revealed that all three AHPs incorporate activation of calcium-gated potassium channels. The ionic basis for a fAHP was rapidly attributed to the actions of big conductance (BK) and the mAHP to small conductance (SK) or Kv7 potassium channels. In stark contrast, the ionic basis for a prominent slow AHP of up to 30 s duration remained an enigma for over 30 years. Recent advances in pharmacological, molecular, and imaging tools have uncovered the expression of a calcium-gated intermediate conductance potassium channel (IK, KCa3.1) in central neurons that proves to contribute to the slow AHP in CA1 hippocampal pyramidal cells. Together the data show that the sAHP arises in part from a core tripartite complex between Cav1.3 (L-type) calcium channels, ryanodine receptors, and IK channels at endoplasmic reticulum-plasma membrane junctions. Work on the sAHP in CA1 pyramidal neurons has again quickened pace, with identified contributions by both IK channels and the Na-K pump providing answers to several mysteries in the pharmacological properties of the sAHP.
Collapse
Affiliation(s)
- Giriraj Sahu
- National Institute of Pharmaceutical Education and Research Ahmedabad, Ahmedabad, India
| | - Ray W Turner
- Department Cell Biology & Anatomy, Cumming School of Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
27
|
Poggio E, Brini M, Calì T. Get Closer to the World of Contact Sites: A Beginner's Guide to Proximity-Driven Fluorescent Probes. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2022; 5:25152564221135748. [PMID: 37366505 PMCID: PMC10243574 DOI: 10.1177/25152564221135748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
To maintain cellular homeostasis and to coordinate the proper response to a specific stimulus, information must be integrated throughout the cell in a well-organized network, in which organelles are the crucial nodes and membrane contact sites are the main edges. Membrane contact sites are the cellular subdomains where two or more organelles come into close apposition and interact with each other. Even though many inter-organelle contacts have been identified, most of them are still not fully characterized, therefore their study is an appealing and expanding field of research. Thanks to significant technological progress, many tools are now available or are in rapid development, making it difficult to choose which one is the most suitable for answering a specific biological question. Here we distinguish two different experimental approaches for studying inter-organelle contact sites. The first one aims to morphologically characterize the sites of membrane contact and to identify the molecular players involved, relying mainly on the application of biochemical and electron microscopy (EM)-related methods. The second approach aims to understand the functional importance of a specific contact, focusing on spatio-temporal details. For this purpose, proximity-driven fluorescent probes are the experimental tools of choice, since they allow the monitoring and quantification of membrane contact sites and their dynamics in living cells under different cellular conditions or upon different stimuli. In this review, we focus on these tools with the purpose of highlighting their great versatility and how they can be applied in the study of membrane contacts. We will extensively describe all the different types of proximity-driven fluorescent tools, discussing their benefits and drawbacks, ultimately providing some suggestions to choose and apply the appropriate methods on a case-to-case basis and to obtain the best experimental outcomes.
Collapse
Affiliation(s)
- Elena Poggio
- Department of Biology (DiBio), University of
Padova, Padova, Italy
| | - Marisa Brini
- Department of Biology (DiBio), University of
Padova, Padova, Italy
- Study Center for Neurodegeneration (CESNE),
University of Padova, Padova, Italy
| | - Tito Calì
- Study Center for Neurodegeneration (CESNE),
University of Padova, Padova, Italy
- Department of Biomedical Sciences (DSB),
University of Padova, Padova, Italy
- Padova Neuroscience Center (PNC), University
of Padova, Padova, Italy
| |
Collapse
|
28
|
Thapa P, Stewart R, Sepela RJ, Vivas O, Parajuli LK, Lillya M, Fletcher-Taylor S, Cohen BE, Zito K, Sack JT. EVAP: A two-photon imaging tool to study conformational changes in endogenous Kv2 channels in live tissues. J Gen Physiol 2021; 153:212666. [PMID: 34581724 PMCID: PMC8480965 DOI: 10.1085/jgp.202012858] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 09/03/2021] [Indexed: 12/29/2022] Open
Abstract
A primary goal of molecular physiology is to understand how conformational changes of proteins affect the function of cells, tissues, and organisms. Here, we describe an imaging method for measuring the conformational changes of the voltage sensors of endogenous ion channel proteins within live tissue, without genetic modification. We synthesized GxTX-594, a variant of the peptidyl tarantula toxin guangxitoxin-1E, conjugated to a fluorophore optimal for two-photon excitation imaging through light-scattering tissue. We term this tool EVAP (Endogenous Voltage-sensor Activity Probe). GxTX-594 targets the voltage sensors of Kv2 proteins, which form potassium channels and plasma membrane–endoplasmic reticulum junctions. GxTX-594 dynamically labels Kv2 proteins on cell surfaces in response to voltage stimulation. To interpret dynamic changes in fluorescence intensity, we developed a statistical thermodynamic model that relates the conformational changes of Kv2 voltage sensors to degree of labeling. We used two-photon excitation imaging of rat brain slices to image Kv2 proteins in neurons. We found puncta of GxTX-594 on hippocampal CA1 neurons that responded to voltage stimulation and retain a voltage response roughly similar to heterologously expressed Kv2.1 protein. Our findings show that EVAP imaging methods enable the identification of conformational changes of endogenous Kv2 voltage sensors in tissue.
Collapse
Affiliation(s)
- Parashar Thapa
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Robert Stewart
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Rebecka J Sepela
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Oscar Vivas
- Center for Neuroscience, University of California, Davis, Davis, CA
| | - Laxmi K Parajuli
- Center for Neuroscience, University of California, Davis, Davis, CA
| | - Mark Lillya
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Sebastian Fletcher-Taylor
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA.,The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Bruce E Cohen
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA.,Division of Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, Davis, CA
| | - Jon T Sack
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA.,Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA
| |
Collapse
|
29
|
Kang Y, Liu J, Jiang Y, Yin S, Huang Z, Zhang Y, Wu J, Chen L, Shao L. Understanding the interactions between inorganic-based nanomaterials and biological membranes. Adv Drug Deliv Rev 2021; 175:113820. [PMID: 34087327 DOI: 10.1016/j.addr.2021.05.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
The interactions between inorganic-based nanomaterials (NMs) and biological membranes are among the most important phenomena for developing NM-based therapeutics and resolving nanotoxicology. Herein, we introduce the structural and functional effects of inorganic-based NMs on biological membranes, mainly the plasma membrane and the endomembrane system, with an emphasis on the interface, which involves highly complex networks between NMs and biomolecules (such as membrane proteins and lipids). Significant efforts have been devoted to categorizing and analyzing the interaction mechanisms in terms of the physicochemical characteristics and biological effects of NMs, which can directly or indirectly influence the effects of NMs on membranes. Importantly, we summarize that the biological membranes act as platforms and thereby mediate NMs-immune system contacts. In this overview, the existing challenges and potential applications in the areas are addressed. A strong understanding of the discussed concepts will promote therapeutic NM designs for drug delivery systems by leveraging the NMs-membrane interactions and their functions.
Collapse
Affiliation(s)
- Yiyuan Kang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China
| | - Jia Liu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanping Jiang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Suhan Yin
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhendong Huang
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanli Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Junrong Wu
- Stomatological Hospital, Southern Medical University, Guangzhou 510280, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Longquan Shao
- Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou 510515, China.
| |
Collapse
|
30
|
Maverick EE, Leek AN, Tamkun MM. Kv2 channel-AMIGO β-subunit assembly modulates both channel function and cell adhesion molecule surface trafficking. J Cell Sci 2021; 134:jcs256339. [PMID: 34137443 PMCID: PMC8255027 DOI: 10.1242/jcs.256339] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 05/17/2021] [Indexed: 11/20/2022] Open
Abstract
The Kv2 channels encode delayed rectifier currents that regulate membrane potential in many tissues. They also have a non-conducting function to form stable junctions between the endoplasmic reticulum and plasma membranes, creating membrane contact sites that mediate functions distinct from membrane excitability. Therefore, proteins that interact with Kv2.1 and Kv2.2 channels can alter conducting and/or non-conducting channel properties. One member of the AMIGO family of proteins is an auxiliary β-subunit for Kv2 channels and modulates Kv2.1 electrical activity. However, the AMIGO family has two additional members of ∼50% similarity that have not yet been characterized as Kv2 β-subunits. In this work, we show that the surface trafficking and localization of all three AMIGOs are controlled by their assembly with both Kv2 channels. Additionally, assembly of each AMIGO with either Kv2.1 or Kv2.2 hyperpolarizes the channel activation midpoint by -10 mV. However, only AMIGO2 significantly slows inactivation and deactivation, leading to a prolonged open state of Kv2 channels. The co-regulatory effects of Kv2s and AMIGOs likely fine-tune both the electrical and non-electrical properties of the cells in which they are expressed.
Collapse
Affiliation(s)
- Emily E. Maverick
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Ashley N. Leek
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523, USA
| | - Michael M. Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO 80523, USA
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
31
|
Lipkin AM, Cunniff MM, Spratt PWE, Lemke SM, Bender KJ. Functional Microstructure of Ca V-Mediated Calcium Signaling in the Axon Initial Segment. J Neurosci 2021; 41:3764-3776. [PMID: 33731449 PMCID: PMC8084313 DOI: 10.1523/jneurosci.2843-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/15/2021] [Accepted: 03/09/2021] [Indexed: 01/12/2023] Open
Abstract
The axon initial segment (AIS) is a specialized neuronal compartment in which synaptic input is converted into action potential (AP) output. This process is supported by a diverse complement of sodium, potassium, and calcium channels (CaV). Different classes of sodium and potassium channels are scaffolded at specific sites within the AIS, conferring unique functions, but how calcium channels are functionally distributed within the AIS is unclear. Here, we use conventional two-photon laser scanning and diffraction-limited, high-speed spot two-photon imaging to resolve AP-evoked calcium dynamics in the AIS with high spatiotemporal resolution. In mouse layer 5 prefrontal pyramidal neurons, calcium influx was mediated by a mix of CaV2 and CaV3 channels that differentially localized to discrete regions. CaV3 functionally localized to produce nanodomain hotspots of calcium influx that coupled to ryanodine-sensitive stores, whereas CaV2 localized to non-hotspot regions. Thus, different pools of CaVs appear to play distinct roles in AIS function.SIGNIFICANCE STATEMENT The axon initial segment (AIS) is the site where synaptic input is transformed into action potential (AP) output. It achieves this function through a diverse complement of sodium, potassium, and calcium channels (CaV). While the localization and function of sodium channels and potassium channels at the AIS is well described, less is known about the functional distribution of CaVs. We used high-speed two-photon imaging to understand activity-dependent calcium dynamics in the AIS of mouse neocortical pyramidal neurons. Surprisingly, we found that calcium influx occurred in two distinct domains: CaV3 generates hotspot regions of calcium influx coupled to calcium stores, whereas CaV2 channels underlie diffuse calcium influx between hotspots. Therefore, different CaV classes localize to distinct AIS subdomains, possibly regulating distinct cellular processes.
Collapse
Affiliation(s)
- Anna M Lipkin
- Neuroscience Graduate Program, University of California, San Francisco, California 94158
| | - Margaret M Cunniff
- Neuroscience Graduate Program, University of California, San Francisco, California 94158
| | - Perry W E Spratt
- Neuroscience Graduate Program, University of California, San Francisco, California 94158
| | - Stefan M Lemke
- Neuroscience Graduate Program, University of California, San Francisco, California 94158
| | - Kevin J Bender
- Neuroscience Graduate Program, University of California, San Francisco, California 94158
- Department of Neurology, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California, San Francisco, California 94158
| |
Collapse
|
32
|
Hewlett B, Singh NP, Vannier C, Galli T. ER-PM Contact Sites - SNARING Actors in Emerging Functions. Front Cell Dev Biol 2021; 9:635518. [PMID: 33681218 PMCID: PMC7928305 DOI: 10.3389/fcell.2021.635518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
The compartmentalisation achieved by confining cytoplasm into membrane-enclosed organelles in eukaryotic cells is essential for maintaining vital functions including ATP production, synthetic and degradative pathways. While intracellular organelles are highly specialised in these functions, the restricting membranes also impede exchange of molecules responsible for the synchronised and responsive cellular activities. The initial identification of contact sites between the ER and plasma membrane (PM) provided a potential candidate structure for communication between organelles without mixing by fusion. Over the past decades, research has revealed a far broader picture of the events. Membrane contact sites (MCSs) have been recognized as increasingly important actors in cell differentiation, plasticity and maintenance, and, upon dysfunction, responsible for pathological conditions such as cancer and neurodegenerative diseases. Present in multiple organelles and cell types, MCSs promote transport of lipids and Ca2+ homoeostasis, with a range of associated protein families. Interestingly, each MCS displays a unique molecular signature, adapted to organelle functions. This review will explore the literature describing the molecular components and interactions taking place at ER-PM contact sites, their functions, and implications in eukaryotic cells, particularly neurons, with emphasis on lipid transfer proteins and emerging function of SNAREs.
Collapse
Affiliation(s)
- Bailey Hewlett
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Neha Pratap Singh
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Christian Vannier
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Thierry Galli
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France.,GHU PARIS Psychiatrie and Neurosciences, Paris, France
| |
Collapse
|
33
|
Li C, Qian T, He R, Wan C, Liu Y, Yu H. Endoplasmic Reticulum-Plasma Membrane Contact Sites: Regulators, Mechanisms, and Physiological Functions. Front Cell Dev Biol 2021; 9:627700. [PMID: 33614657 PMCID: PMC7889955 DOI: 10.3389/fcell.2021.627700] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) forms direct membrane contact sites with the plasma membrane (PM) in eukaryotic cells. These ER-PM contact sites play essential roles in lipid homeostasis, ion dynamics, and cell signaling, which are carried out by protein-protein or protein-lipid interactions. Distinct tethering factors dynamically control the architecture of ER-PM junctions in response to intracellular signals or external stimuli. The physiological roles of ER-PM contact sites are dependent on a variety of regulators that individually or cooperatively perform functions in diverse cellular processes. This review focuses on proteins functioning at ER-PM contact sites and highlights the recent progress in their mechanisms and physiological roles.
Collapse
Affiliation(s)
- Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
34
|
Deardorff AS, Romer SH, Fyffe RE. Location, location, location: the organization and roles of potassium channels in mammalian motoneurons. J Physiol 2021; 599:1391-1420. [DOI: 10.1113/jp278675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/08/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Adam S. Deardorff
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
- Department of Neurology and Internal Medicine, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| | - Shannon H. Romer
- Odyssey Systems Environmental Health Effects Laboratory, Navy Medical Research Unit‐Dayton Wright‐Patterson Air Force Base OH 45433 USA
| | - Robert E.W. Fyffe
- Department of Neuroscience, Cell Biology and Physiology, Wright State University Boonshoft School of Medicine Dayton OH 45435 USA
| |
Collapse
|
35
|
Farah A, Kabbage M, Atafi S, Gabteni AJ, Barbirou M, Madhioub M, Hamzaoui L, Mohamed MA, Touinsi H, Kchaou AO, Chelbi E, Boubaker S, Abderrazek RB, Bouhaouala-Zahar B. Selective expression of KCNA5 and KCNB1 genes in gastric and colorectal carcinoma. BMC Cancer 2020; 20:1179. [PMID: 33267786 PMCID: PMC7709444 DOI: 10.1186/s12885-020-07647-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 11/17/2020] [Indexed: 01/04/2023] Open
Abstract
Background Gastric and colorectal cancers are the most common malignant tumours, leading to a significant number of cancer-related deaths worldwide. Recently, increasing evidence has demonstrated that cancer cells exhibit a differential expression of potassium channels and this can contribute to cancer progression. However, their expression and localisation at the somatic level remains uncertain. In this study, we have investigated the expression levels of KCNB1 and KCNA5 genes encoding ubiquitous Kv2.1 and Kv1.5 potassium channels in gastric and colorectal tumours. Methods Gastric and colorectal tumoral and peritumoral tissues were collected to evaluate the expression of KCNB1 and KCNA5 mRNA by quantitative PCR. Moreover, the immunohistochemical staining profile of Kv2.1 and Kv1.5 was assessed on 40 Formalin-Fixed and Paraffin-Embedded (FFPE) gastric carcinoma tissues. Differences in gene expression between tumoral and peritumoral tissues were compared statistically with the Mann-Whitney U test. The association between the clinicopathological features of the GC patients and the expression of both Kv proteins was investigated with χ2 and Fisher’s exact tests. Results The mRNA fold expression of KCNB1 and KCNA5 genes showed a lower mean in the tumoral tissues (0.06 ± 0.17, 0.006 ± 0.009) compared to peritumoral tissues (0.08 ± 0.16, 0.16 ± 0.48, respectively) without reaching the significance rate (p = 0.861, p = 0.152, respectively). Interestingly, Kv2.1 and Kv1.5 immunostaining was detectable and characterised by a large distribution in peritumoral and tumoral epithelial cells. More interestingly, inflammatory cells were also stained. Surprisingly, Kv2.1 and Kv1.5 staining was undoubtedly and predominantly detected in the cytoplasm compartment of tumour cells. Indeed, the expression of Kv2.1 in tumour cells revealed a significant association with the early gastric cancer clinical stage (p = 0.026). Conclusion The data highlight, for the first time, the potential role of Kv1.5 and Kv2.1 in gastrointestinal-related cancers and suggests they may be promising prognostic markers for these tumours.
Collapse
Affiliation(s)
- Azer Farah
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia
| | - Maria Kabbage
- Biomedical Genomics and Oncogenetics Laboratory, LR11IPT05 Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Salsabil Atafi
- Laboratory of Human and Experimental Pathology, Institute Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Amira Jaballah Gabteni
- Biomedical Genomics and Oncogenetics Laboratory, LR11IPT05 Institut Pasteur de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Mouadh Barbirou
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia.,Center for Biomedical Informatics, Department of Health Management and Informatics, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mouna Madhioub
- Gastroenterology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | - Lamine Hamzaoui
- Gastroenterology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | | | - Hassen Touinsi
- Surgical Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | | | - Emna Chelbi
- Pathology Department, Mohamed Tahar Maamouri Hospital, 8000, Nabeul, Tunisia
| | - Samir Boubaker
- Laboratory of Human and Experimental Pathology, Institute Pasteur Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rahma Ben Abderrazek
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia.
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Biomolecules, LR16IPT08 Institute Pasteur Tunis, Tunis Belvédère- University of Tunis El Manar, 13 Place Pasteur, BP74, Tunis, Tunisia. .,Medical School of Tunis, University of Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
36
|
Di Mattia T, Martinet A, Ikhlef S, McEwen AG, Nominé Y, Wendling C, Poussin-Courmontagne P, Voilquin L, Eberling P, Ruffenach F, Cavarelli J, Slee J, Levine TP, Drin G, Tomasetto C, Alpy F. FFAT motif phosphorylation controls formation and lipid transfer function of inter-organelle contacts. EMBO J 2020; 39:e104369. [PMID: 33124732 PMCID: PMC7705450 DOI: 10.15252/embj.2019104369] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 11/13/2022] Open
Abstract
Organelles are physically connected in membrane contact sites. The endoplasmic reticulum possesses three major receptors, VAP‐A, VAP‐B, and MOSPD2, which interact with proteins at the surface of other organelles to build contacts. VAP‐A, VAP‐B, and MOSPD2 contain an MSP domain, which binds a motif named FFAT (two phenylalanines in an acidic tract). In this study, we identified a non‐conventional FFAT motif where a conserved acidic residue is replaced by a serine/threonine. We show that phosphorylation of this serine/threonine is critical for non‐conventional FFAT motifs (named Phospho‐FFAT) to be recognized by the MSP domain. Moreover, structural analyses of the MSP domain alone or in complex with conventional and Phospho‐FFAT peptides revealed new mechanisms of interaction. Based on these new insights, we produced a novel prediction algorithm, which expands the repertoire of candidate proteins with a Phospho‐FFAT that are able to create membrane contact sites. Using a prototypical tethering complex made by STARD3 and VAP, we showed that phosphorylation is instrumental for the formation of ER‐endosome contacts, and their sterol transfer function. This study reveals that phosphorylation acts as a general switch for inter‐organelle contacts.
Collapse
Affiliation(s)
- Thomas Di Mattia
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Arthur Martinet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Souade Ikhlef
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, Valbonne, France
| | - Alastair G McEwen
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Yves Nominé
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Corinne Wendling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Pierre Poussin-Courmontagne
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Laetitia Voilquin
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Pascal Eberling
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Frank Ruffenach
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Jean Cavarelli
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - John Slee
- UCL Institute of Ophthalmology, London, UK
| | | | - Guillaume Drin
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, CNRS, Valbonne, France
| | - Catherine Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Fabien Alpy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, Illkirch, France.,Centre National de la Recherche Scientifique (CNRS), UMR 7104, Illkirch, France.,Université de Strasbourg, Illkirch, France
| |
Collapse
|
37
|
Hawkins NA, Misra SN, Jurado M, Kang SK, Vierra NC, Nguyen K, Wren L, George AL, Trimmer JS, Kearney JA. Epilepsy and neurobehavioral abnormalities in mice with a dominant-negative KCNB1 pathogenic variant. Neurobiol Dis 2020; 147:105141. [PMID: 33132203 PMCID: PMC7725922 DOI: 10.1016/j.nbd.2020.105141] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 11/23/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEE) are a group of severe epilepsies that usually present with intractable seizures, developmental delay, and often have elevated risk for premature mortality. Numerous genes have been identified as a monogenic cause of DEE, including KCNB1. The voltage-gated potassium channel Kv2.1, encoded by KCNB1, is primarily responsible for delayed rectifier potassium currents that are important regulators of excitability in electrically excitable cells, including neurons. In addition to its canonical role as a voltage-gated potassium conductance, Kv2.1 also serves a highly conserved structural function organizing endoplasmic reticulum-plasma membrane junctions clustered in the soma and proximal dendrites of neurons. The de novo pathogenic variant KCNB1-p.G379R was identified in an infant with epileptic spasms, and atonic, focal and tonic-clonic seizures that were refractory to treatment with standard antiepileptic drugs. Previous work demonstrated deficits in potassium conductance, but did not assess non-conducting functions. To determine if the G379R variant affected Kv2.1 clustering at endoplasmic reticulum-plasma membrane junctions, Kv2.1-G379R was expressed in HEK293T cells. Kv2.1-G379R expression did not induce formation of endoplasmic reticulum-plasma membrane junctions, and co-expression of Kv2.1-G379R with Kv2.1-wild-type lowered induction of these structures relative to Kv2.1-WT alone, consistent with a dominant negative effect. To model this variant in vivo, we introduced Kcnb1G379R into mice using CRISPR/Cas9 genome editing. We characterized neuronal expression, neurological and neurobehavioral phenotypes of Kcnb1G379R/+ (Kcnb1R/+) and Kcnb1G379R/G379R (Kcnb1R/R) mice. Immunohistochemistry studies on brains from Kcnb1+/+, Kcnb1R/+ and Kcnb1R/R mice revealed genotype-dependent differences in the expression levels of Kv2.1 protein, as well as associated Kv2.2 and AMIGO-1 proteins. Kcnb1R/+ and Kcnb1R/R mice displayed profound hyperactivity, repetitive behaviors, impulsivity and reduced anxiety. Spontaneous seizures were observed in Kcnb1R/R mice, as well as seizures induced by exposure to novel environments and/ or handling. Both Kcnb1R/+ and Kcnb1R/R mutants were more susceptible to proconvulsant-induced seizures. In addition, both Kcnb1R/+ and Kcnb1R/R mice exhibited abnormal interictal EEG activity, including isolated spike and slow waves. Overall, the Kcnb1G379R mice recapitulate many features observed in individuals with DEE due to pathogenic variants in KCNB1. This new mouse model of KCNB1-associated DEE will be valuable for improving the understanding of the underlying pathophysiology and will provide a valuable tool for the development of therapies to treat this pharmacoresistant DEE.
Collapse
Affiliation(s)
- Nicole A Hawkins
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Sunita N Misra
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America; Ann & Robert H. Lurie Children's Hospital of Chicago Chicago, IL 60611, United States of America
| | - Manuel Jurado
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Seok Kyu Kang
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Nicholas C Vierra
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616, United States of America; Department of preceding Physiology and Membrane Biology, University of California, Davis, CA 95616, United States of America
| | - Kimberly Nguyen
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616, United States of America
| | - Lisa Wren
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - Alfred L George
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95616, United States of America; Department of preceding Physiology and Membrane Biology, University of California, Davis, CA 95616, United States of America
| | - Jennifer A Kearney
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States of America.
| |
Collapse
|
38
|
Solé L, Wagnon JL, Tamkun MM. Functional analysis of three Na v1.6 mutations causing early infantile epileptic encephalopathy. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165959. [PMID: 32916281 DOI: 10.1016/j.bbadis.2020.165959] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/21/2020] [Accepted: 09/03/2020] [Indexed: 11/24/2022]
Abstract
The voltage-gated sodium channel Nav1.6 is associated with more than 300 cases of epileptic encephalopathy. Nav1.6 epilepsy-causing mutations are spread over the entire channel's structure and only 10% of mutations have been characterized at the molecular level, with most of them being gain of function mutations. In this study, we analyzed three previously uncharacterized Nav1.6 epilepsy-causing mutations: G214D, N215D and V216D, located within a mutation hot-spot at the S3-S4 extracellular loop of Domain1. Voltage clamp experiments showed a 6-16 mV hyperpolarizing shift in the activation mid-point for all three mutants. V216D presented the largest shift along with decreased current amplitude, enhanced inactivation and a lack of persistent current. Recordings at hyperpolarized potentials indicated that all three mutants presented gating pore currents. Furthermore, trafficking experiments performed in cultured hippocampal neurons demonstrated that the mutants trafficked properly to the cell surface, with no significant differences regarding surface expression within the axon initial segment or soma compared to wild-type. These trafficking data suggest that the disease-causing consequences are due to only changes in the biophysical properties of the channel. Interestingly, the patient carrying the V216D mutation, which is the mutant with the greatest electrophysiological changes as compared to wild-type, exhibited the most severe phenotype. These results emphasize that these mutations will mandate unique treatment approaches, for normal sodium channel blockers may not work given that the studied mutations present gating pore currents. This study emphasizes the importance of molecular characterization of disease-causing mutations in order to improve the pharmacological treatment of patients.
Collapse
Affiliation(s)
- Laura Solé
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO 80523, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jacy L Wagnon
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Michael M Tamkun
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO 80523, USA; Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
39
|
Yeh CY, Schulien AJ, Molyneaux BJ, Aizenman E. Lessons from Recent Advances in Ischemic Stroke Management and Targeting Kv2.1 for Neuroprotection. Int J Mol Sci 2020; 21:ijms21176107. [PMID: 32854248 PMCID: PMC7503403 DOI: 10.3390/ijms21176107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
Achieving neuroprotection in ischemic stroke patients has been a multidecade medical challenge. Numerous clinical trials were discontinued in futility and many were terminated in response to deleterious treatment effects. Recently, however, several positive reports have generated the much-needed excitement surrounding stroke therapy. In this review, we describe the clinical studies that significantly expanded the time window of eligibility for patients to receive mechanical endovascular thrombectomy. We further summarize the results available thus far for nerinetide, a promising neuroprotective agent for stroke treatment. Lastly, we reflect upon aspects of these impactful trials in our own studies targeting the Kv2.1-mediated cell death pathway in neurons for neuroprotection. We argue that recent changes in the clinical landscape should be adapted by preclinical research in order to continue progressing toward the development of efficacious neuroprotective therapies for ischemic stroke.
Collapse
Affiliation(s)
- Chung-Yang Yeh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.-Y.Y.); (A.J.S.)
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Anthony J. Schulien
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.-Y.Y.); (A.J.S.)
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Bradley J. Molyneaux
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- UPMC Stroke Institute, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (C.-Y.Y.); (A.J.S.)
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- Correspondence:
| |
Collapse
|
40
|
ORP/Osh mediate cross-talk between ER-plasma membrane contact site components and plasma membrane SNAREs. Cell Mol Life Sci 2020; 78:1689-1708. [PMID: 32734583 PMCID: PMC7904734 DOI: 10.1007/s00018-020-03604-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023]
Abstract
OSBP-homologous proteins (ORPs, Oshp) are lipid binding/transfer proteins. Several ORP/Oshp localize to membrane contacts between the endoplasmic reticulum (ER) and the plasma membrane, where they mediate lipid transfer or regulate lipid-modifying enzymes. A common way in which they target contacts is by binding to the ER proteins, VAP/Scs2p, while the second membrane is targeted by other interactions with lipids or proteins.We have studied the cross-talk of secretory SNARE proteins and their regulators with ORP/Oshp and VAPA/Scs2p at ER-plasma membrane contact sites in yeast and murine primary neurons. We show that Oshp-Scs2p interactions depend on intact secretory SNARE proteins, especially Sec9p. SNAP-25/Sec9p directly interact with ORP/Osh proteins and their disruption destabilized the ORP/Osh proteins, associated with dysfunction of VAPA/Scs2p. Deleting OSH1-3 in yeast or knocking down ORP2 in primary neurons reduced the oligomerization of VAPA/Scs2p and affected their multiple interactions with SNAREs. These observations reveal a novel cross-talk between the machineries of ER-plasma membrane contact sites and those driving exocytosis.
Collapse
|
41
|
Schulien AJ, Yeh CY, Orange BN, Pav OJ, Hopkins MP, Moutal A, Khanna R, Sun D, Justice JA, Aizenman E. Targeted disruption of Kv2.1-VAPA association provides neuroprotection against ischemic stroke in mice by declustering Kv2.1 channels. SCIENCE ADVANCES 2020; 6:eaaz8110. [PMID: 32937450 PMCID: PMC7458461 DOI: 10.1126/sciadv.aaz8110] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 05/15/2020] [Indexed: 05/07/2023]
Abstract
Kv2.1 channels mediate cell death-enabling loss of cytosolic potassium in neurons following plasma membrane insertion at somatodendritic clusters. Overexpression of the carboxyl terminus (CT) of the cognate channel Kv2.2 is neuroprotective by disrupting Kv2.1 surface clusters. Here, we define a seven-amino acid declustering domain within Kv2.2 CT (DP-2) and demonstrate its neuroprotective efficacy in a murine ischemia-reperfusion model. TAT-DP-2, a membrane-permeable derivative, induces Kv2.1 surface cluster dispersal, prevents post-injurious pro-apoptotic potassium current enhancement, and is neuroprotective in vitro by disrupting the association of Kv2.1 with VAPA. TAT-DP-2 also induces Kv2.1 cluster dispersal in vivo in mice, reducing infarct size and improving long-term neurological function following stroke. We suggest that TAT-DP-2 induces Kv2.1 declustering by disrupting Kv2.1-VAPA association and scaffolding sites required for the membrane insertion of Kv2.1 channels following injury. We present the first evidence of targeted disruption of Kv2.1-VAPA association as a neuroprotective strategy following brain ischemia.
Collapse
Affiliation(s)
- Anthony J Schulien
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chung-Yang Yeh
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Bailey N Orange
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Olivia J Pav
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Madelynn P Hopkins
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Dandan Sun
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jason A Justice
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Elias Aizenman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
42
|
Endoplasmic reticulum-plasma membrane contacts: Principals of phosphoinositide and calcium signaling. Curr Opin Cell Biol 2020; 63:125-134. [PMID: 32088611 DOI: 10.1016/j.ceb.2020.01.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/29/2022]
Abstract
The endoplasmic reticulum (ER) forms an extensive network of membrane contact sites with intra-cellular organelles and the plasma membrane (PM). Interorganelle contacts have vital roles in membrane lipid and ion dynamics. In particular, ER-PM contacts are integral to numerous inter-cellular and intra-cellular signaling pathways including phosphoinositide lipid and calcium signaling, mechanotransduction, metabolic regulation, and cell stress responses. Accordingly, ER-PM contacts serve important signaling functions in excitable cells including neurons and muscle and endocrine cells. This review highlights recent advances in our understanding of the vital roles for ER-PM contacts in phosphoinositide and calcium signaling and how signaling pathways in turn regulate proteins that form and function at ER-PM contacts.
Collapse
|
43
|
Kv2.1 channels play opposing roles in regulating membrane potential, Ca 2+ channel function, and myogenic tone in arterial smooth muscle. Proc Natl Acad Sci U S A 2020; 117:3858-3866. [PMID: 32015129 PMCID: PMC7035623 DOI: 10.1073/pnas.1917879117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The accepted role of the protein Kv2.1 in arterial smooth muscle cells is to form K+ channels in the sarcolemma. Opening of Kv2.1 channels causes membrane hyperpolarization, which decreases the activity of L-type CaV1.2 channels, lowering intracellular Ca2+ ([Ca2+]i) and causing smooth muscle relaxation. A limitation of this model is that it is based exclusively on data from male arterial myocytes. Here, we used a combination of electrophysiology as well as imaging approaches to investigate the role of Kv2.1 channels in male and female arterial myocytes. We confirmed that Kv2.1 plays a canonical conductive role but found it also has a structural role in arterial myocytes to enhance clustering of CaV1.2 channels. Less than 1% of Kv2.1 channels are conductive and induce membrane hyperpolarization. Paradoxically, by enhancing the structural clustering and probability of CaV1.2-CaV1.2 interactions within these clusters, Kv2.1 increases Ca2+ influx. These functional impacts of Kv2.1 depend on its level of expression, which varies with sex. In female myocytes, where expression of Kv2.1 protein is higher than in male myocytes, Kv2.1 has conductive and structural roles. Female myocytes have larger CaV1.2 clusters, larger [Ca2+]i, and larger myogenic tone than male myocytes. In contrast, in male myocytes, Kv2.1 channels regulate membrane potential but not CaV1.2 channel clustering. We propose a model in which Kv2.1 function varies with sex: in males, Kv2.1 channels control membrane potential but, in female myocytes, Kv2.1 plays dual electrical and CaV1.2 clustering roles. This contributes to sex-specific regulation of excitability, [Ca2+]i, and myogenic tone in arterial myocytes.
Collapse
|
44
|
Öztürk Z, O’Kane CJ, Pérez-Moreno JJ. Axonal Endoplasmic Reticulum Dynamics and Its Roles in Neurodegeneration. Front Neurosci 2020; 14:48. [PMID: 32116502 PMCID: PMC7025499 DOI: 10.3389/fnins.2020.00048] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
The physical continuity of axons over long cellular distances poses challenges for their maintenance. One organelle that faces this challenge is endoplasmic reticulum (ER); unlike other intracellular organelles, this forms a physically continuous network throughout the cell, with a single membrane and a single lumen. In axons, ER is mainly smooth, forming a tubular network with occasional sheets or cisternae and low amounts of rough ER. It has many potential roles: lipid biosynthesis, glucose homeostasis, a Ca2+ store, protein export, and contacting and regulating other organelles. This tubular network structure is determined by ER-shaping proteins, mutations in some of which are causative for neurodegenerative disorders such as hereditary spastic paraplegia (HSP). While axonal ER shares many features with the tubular ER network in other contexts, these features must be adapted to the long and narrow dimensions of axons. ER appears to be physically continuous throughout axons, over distances that are enormous on a subcellular scale. It is therefore a potential channel for long-distance or regional communication within neurons, independent of action potentials or physical transport of cargos, but involving its physiological roles such as Ca2+ or organelle homeostasis. Despite its apparent stability, axonal ER is highly dynamic, showing features like anterograde and retrograde transport, potentially reflecting continuous fusion and breakage of the network. Here we discuss the transport processes that must contribute to this dynamic behavior of ER. We also discuss the model that these processes underpin a homeostatic process that ensures both enough ER to maintain continuity of the network and repair breaks in it, but not too much ER that might disrupt local cellular physiology. Finally, we discuss how failure of ER organization in axons could lead to axon degenerative diseases, and how a requirement for ER continuity could make distal axons most susceptible to degeneration in conditions that disrupt ER continuity.
Collapse
Affiliation(s)
| | - Cahir J. O’Kane
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
45
|
Cserép C, Pósfai B, Lénárt N, Fekete R, László ZI, Lele Z, Orsolits B, Molnár G, Heindl S, Schwarcz AD, Ujvári K, Környei Z, Tóth K, Szabadits E, Sperlágh B, Baranyi M, Csiba L, Hortobágyi T, Maglóczky Z, Martinecz B, Szabó G, Erdélyi F, Szipőcs R, Tamkun MM, Gesierich B, Duering M, Katona I, Liesz A, Tamás G, Dénes Á. Microglia monitor and protect neuronal function through specialized somatic purinergic junctions. Science 2019; 367:528-537. [PMID: 31831638 DOI: 10.1126/science.aax6752] [Citation(s) in RCA: 423] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/14/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
Abstract
Microglia are the main immune cells in the brain and have roles in brain homeostasis and neurological diseases. Mechanisms underlying microglia-neuron communication remain elusive. Here, we identified an interaction site between neuronal cell bodies and microglial processes in mouse and human brain. Somatic microglia-neuron junctions have a specialized nanoarchitecture optimized for purinergic signaling. Activity of neuronal mitochondria was linked with microglial junction formation, which was induced rapidly in response to neuronal activation and blocked by inhibition of P2Y12 receptors. Brain injury-induced changes at somatic junctions triggered P2Y12 receptor-dependent microglial neuroprotection, regulating neuronal calcium load and functional connectivity. Thus, microglial processes at these junctions could potentially monitor and protect neuronal functions.
Collapse
Affiliation(s)
- Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Rebeka Fekete
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Zsófia I László
- Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary.,Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zsolt Lele
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Barbara Orsolits
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Molnár
- MTA-SZTE Research Group for Cortical Microcircuits of the Hungarian Academy of Sciences, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Steffanie Heindl
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University, Munich, Germany
| | - Anett D Schwarcz
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Katinka Ujvári
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Krisztina Tóth
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.,Szentágothai János Doctoral School of Neuroscience, Semmelweis University, Budapest, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Mária Baranyi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - László Csiba
- MTA-DE Cerebrovascular and Neurodegenerative Research Group, Department of Neurology, University of Debrecen, Debrecen, Hungary
| | - Tibor Hortobágyi
- Institute of Pathology, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,Centre for Age-Related Medicine, SESAM, Stavanger University Hospital, Stavanger, Norway
| | - Zsófia Maglóczky
- Human Brain Research Laboratory, Institute of Experimental Medicine, Budapest, Hungary
| | - Bernadett Martinecz
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Gábor Szabó
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest, Hungary
| | - Ferenc Erdélyi
- Medical Gene Technology Unit, Institute of Experimental Medicine, Budapest, Hungary
| | - Róbert Szipőcs
- Institute for Solid State Physics and Optics of Wigner RCP, Budapest, Hungary
| | - Michael M Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University, Munich, Germany
| | - Marco Duering
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits of the Hungarian Academy of Sciences, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
46
|
Vierra NC, Kirmiz M, van der List D, Santana LF, Trimmer JS. Kv2.1 mediates spatial and functional coupling of L-type calcium channels and ryanodine receptors in mammalian neurons. eLife 2019; 8:49953. [PMID: 31663850 PMCID: PMC6839919 DOI: 10.7554/elife.49953] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/29/2019] [Indexed: 12/16/2022] Open
Abstract
The voltage-gated K+ channel Kv2.1 serves a major structural role in the soma and proximal dendrites of mammalian brain neurons, tethering the plasma membrane (PM) to endoplasmic reticulum (ER). Although Kv2.1 clustering at neuronal ER-PM junctions (EPJs) is tightly regulated and highly conserved, its function remains unclear. By identifying and evaluating proteins in close spatial proximity to Kv2.1-containing EPJs, we discovered that a significant role of Kv2.1 at EPJs is to promote the clustering and functional coupling of PM L-type Ca2+ channels (LTCCs) to ryanodine receptor (RyR) ER Ca2+ release channels. Kv2.1 clustering also unexpectedly enhanced LTCC opening at polarized membrane potentials. This enabled Kv2.1-LTCC-RyR triads to generate localized Ca2+ release events (i.e., Ca2+ sparks) independently of action potentials. Together, these findings uncover a novel mode of LTCC regulation and establish a unique mechanism whereby Kv2.1-associated EPJs provide a molecular platform for localized somatodendritic Ca2+ signals in mammalian brain neurons.
Collapse
Affiliation(s)
- Nicholas C Vierra
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, United States.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, United States
| | - Michael Kirmiz
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, United States
| | - Deborah van der List
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, United States.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, United States
| | - James S Trimmer
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, United States.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, United States
| |
Collapse
|
47
|
Fowler PC, Garcia-Pardo ME, Simpson JC, O'Sullivan NC. NeurodegenERation: The Central Role for ER Contacts in Neuronal Function and Axonopathy, Lessons From Hereditary Spastic Paraplegias and Related Diseases. Front Neurosci 2019; 13:1051. [PMID: 31680803 PMCID: PMC6801308 DOI: 10.3389/fnins.2019.01051] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/19/2019] [Indexed: 12/17/2022] Open
Abstract
The hereditary spastic paraplegias (HSPs) are a group of inherited neurodegenerative conditions whose characteristic feature is degeneration of the longest axons within the corticospinal tract which leads to progressive spasticity and weakness of the lower limbs. Though highly genetically heterogeneous, the majority of HSP cases are caused by mutations in genes encoding proteins that are responsible for generating and organizing the tubular endoplasmic reticulum (ER). Despite this, the role of the ER within neurons, particularly the long axons affected in HSP, is not well understood. Throughout axons, ER tubules make extensive contacts with other organelles, the cytoskeleton and the plasma membrane. At these ER contacts, protein complexes work in concert to perform specialized functions including organelle shaping, calcium homeostasis and lipid biogenesis, all of which are vital for neuronal survival and may be disrupted by HSP-causing mutations. In this article we summarize the proteins which mediate ER contacts, review the functions these contacts are known to carry out within neurons, and discuss the potential contribution of disruption of ER contacts to axonopathy in HSP.
Collapse
Affiliation(s)
- Philippa C Fowler
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - M Elena Garcia-Pardo
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Jeremy C Simpson
- UCD School of Biology and Environmental Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Niamh C O'Sullivan
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
48
|
Jędrychowska J, Korzh V. Kv2.1 voltage-gated potassium channels in developmental perspective. Dev Dyn 2019; 248:1180-1194. [PMID: 31512327 DOI: 10.1002/dvdy.114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 11/11/2022] Open
Abstract
Kv2.1 voltage-gated potassium channels consist of two types of α-subunits: (a) electrically-active Kcnb1 α-subunits and (b) silent or modulatory α-subunits plus β-subunits that, similar to silent α-subunits, also regulate electrically-active subunits. Voltage-gated potassium channels were traditionally viewed, mainly by electrophysiologists, as regulators of the electrical activity of the plasma membrane in excitable cells, a role that is performed by transmembrane protein domains of α-subunits that form the electric pore. Genetic studies revealed a role for this region of α-subunits of voltage-gated potassium channels in human neurodevelopmental disorders, such as epileptic encephalopathy. The N- and C-terminal domains of α-subunits interact to form the cytoplasmic subunit of heterotetrameric potassium channels that regulate electric pores. Subsequent animal studies revealed the developmental functions of Kcnb1-containing voltage-gated potassium channels and illustrated their role during brain development and reproduction. These functions of potassium channels are discussed in this review in the context of regulatory interactions between electrically-active and regulatory subunits.
Collapse
Affiliation(s)
- Justyna Jędrychowska
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Vladimir Korzh
- International Institute of Molecular and Cell Biology in Warsaw, Warsaw, Poland
| |
Collapse
|
49
|
Kirmiz M, Gillies TE, Dickson EJ, Trimmer JS. Neuronal ER-plasma membrane junctions organized by Kv2-VAP pairing recruit Nir proteins and affect phosphoinositide homeostasis. J Biol Chem 2019; 294:17735-17757. [PMID: 31594866 DOI: 10.1074/jbc.ra119.007635] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 10/02/2019] [Indexed: 12/21/2022] Open
Abstract
The association of plasma membrane (PM)-localized voltage-gated potassium (Kv2) channels with endoplasmic reticulum (ER)-localized vesicle-associated membrane protein-associated proteins VAPA and VAPB defines ER-PM junctions in mammalian brain neurons. Here, we used proteomics to identify proteins associated with Kv2/VAP-containing ER-PM junctions. We found that the VAP-interacting membrane-associated phosphatidylinositol (PtdIns) transfer proteins PYK2 N-terminal domain-interacting receptor 2 (Nir2) and Nir3 specifically associate with Kv2.1 complexes. When coexpressed with Kv2.1 and VAPA in HEK293T cells, Nir2 colocalized with cell-surface-conducting and -nonconducting Kv2.1 isoforms. This was enhanced by muscarinic-mediated PtdIns(4,5)P2 hydrolysis, leading to dynamic recruitment of Nir2 to Kv2.1 clusters. In cultured rat hippocampal neurons, exogenously expressed Nir2 did not strongly colocalize with Kv2.1, unless exogenous VAPA was also expressed, supporting the notion that VAPA mediates the spatial association of Kv2.1 and Nir2. Immunolabeling signals of endogenous Kv2.1, Nir2, and VAP puncta were spatially correlated in cultured neurons. Fluorescence-recovery-after-photobleaching experiments revealed that Kv2.1, VAPA, and Nir2 have comparable turnover rates at ER-PM junctions, suggesting that they form complexes at these sites. Exogenous Kv2.1 expression in HEK293T cells resulted in significant differences in the kinetics of PtdIns(4,5)P2 recovery following repetitive muscarinic stimulation, with no apparent impact on resting PtdIns(4,5)P2 or PtdIns(4)P levels. Finally, the brains of Kv2.1-knockout mice had altered composition of PtdIns lipids, suggesting a crucial role for native Kv2.1-containing ER-PM junctions in regulating PtdIns lipid metabolism in brain neurons. These results suggest that ER-PM junctions formed by Kv2 channel-VAP pairing regulate PtdIns lipid homeostasis via VAP-associated PtdIns transfer proteins.
Collapse
Affiliation(s)
- Michael Kirmiz
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California 95616
| | - Taryn E Gillies
- Department of Bioengineering, Stanford University, Stanford, California 94305
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, California 95616
| | - James S Trimmer
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California 95616 .,Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, California 95616
| |
Collapse
|
50
|
Johnson B, Leek AN, Tamkun MM. Kv2 channels create endoplasmic reticulum / plasma membrane junctions: a brief history of Kv2 channel subcellular localization. Channels (Austin) 2019; 13:88-101. [PMID: 30712450 PMCID: PMC6380216 DOI: 10.1080/19336950.2019.1568824] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The potassium channels Kv2.1 and Kv2.2 are widely expressed throughout the mammalian brain. Kv2.1 provides the majority of delayed rectifying current in rat hippocampus while both channels are differentially expressed in cortex. Particularly unusual is their neuronal surface localization pattern: while half the channel population is freely-diffusive on the plasma membrane as expected from the generalized Singer & Nicolson fluid mosaic model, the other half localizes into micron-sized clusters on the soma, dendrites, and axon initial segment. These clusters contain hundreds of channels, which for Kv2.1, are largely non-conducting. Competing theories of the mechanism underlying Kv2.1 clustering have included static tethering to being corralled by an actin fence. Now, recent work has demonstrated channel clustering is due to formation of endoplasmic reticulum/plasma membrane (ER/PM) junctions through interaction with ER-resident VAMP-associated proteins (VAPs). Interaction between surface Kv2 channels and ER VAPs groups channels together in clusters. ER/PM junctions play important roles in inter-organelle communication: they regulate ion flux, are involved in lipid transfer, and are sites of endo- and exocytosis. Kv2-induced ER/PM junctions are regulated through phosphorylation of the channel C-terminus which in turn regulates VAP binding, providing a rapid means to create or dismantle these microdomains. In addition, insults such as hypoxia or ischemia disrupt this interaction resulting in ER/PM junction disassembly. Kv2 channels are the only known plasma membrane protein to form regulated, injury sensitive junctions in this manner. Furthermore, it is likely that concentrated VAPs at these microdomains sequester additional interactors whose functions are not yet fully understood.
Collapse
Affiliation(s)
- Ben Johnson
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Ashley N Leek
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Michael M Tamkun
- a Molecular, Cellular and Integrative Neurosciences Graduate Program , Colorado State University , Fort Collins , CO , USA.,b Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA.,c Department of Biochemistry and Molecular Biology , Colorado State University , Fort Collins , CO , USA
| |
Collapse
|