1
|
Tyagi S, Higerd-Rusli GP, Akin EJ, Waxman SG, Dib-Hajj SD. Sculpting excitable membranes: voltage-gated ion channel delivery and distribution. Nat Rev Neurosci 2025; 26:313-332. [PMID: 40175736 DOI: 10.1038/s41583-025-00917-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2025] [Indexed: 04/04/2025]
Abstract
The polarized and domain-specific distribution of membrane ion channels is essential for neuronal homeostasis, but delivery of these proteins to distal neuronal compartments (such as the axonal ends of peripheral sensory neurons) presents a logistical challenge. Recent developments have enabled the real-time imaging of single protein trafficking and the investigation of the life cycle of ion channels across neuronal compartments. These studies have revealed a highly regulated process involving post-translational modifications, vesicular sorting, motor protein-driven transport and targeted membrane insertion. Emerging evidence suggests that neuronal activity and disease states can dynamically modulate ion channel localization, directly influencing excitability. This Review synthesizes current knowledge on the spatiotemporal regulation of ion channel trafficking in both central and peripheral nervous system neurons. Understanding these processes not only advances our fundamental knowledge of neuronal excitability, but also reveals potential therapeutic targets for disorders involving aberrant ion channel distribution, such as chronic pain and neurodegenerative diseases.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, West Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA.
| | - Grant P Higerd-Rusli
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, West Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Elizabeth J Akin
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, West Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Neuroscience and Regeneration Research, Yale School of Medicine, West Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
2
|
Oldre EN, Webb BD, Sperringer JE, Maness PF. Regulation of perisomatic synapses from cholecystokinin basket interneurons through NrCAM and Ankyrin B. CURRENT RESEARCH IN NEUROBIOLOGY 2025; 8:100150. [PMID: 40276719 PMCID: PMC12018208 DOI: 10.1016/j.crneur.2025.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/07/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
The perisomatic region of cortical pyramidal neurons (PNs) integrates local and long-range inputs and regulates firing. This domain receives GABAergic inputs from cholecystokinin (CCK)- and Parvalbumin (PV)-expressing basket cells (BCs) but how synaptic contacts are established is unclear. Neuron-glial related cell adhesion molecule (NrCAM) is a homophilic transmembrane protein that binds the scaffold protein Ankyrin B. Here we show that NrCAM and Ankyrin B mediate perisomatic synaptic contact between CCK-BCs and PNs in mouse medial prefrontal cortex (mPFC). Immunolabeling of CCK-BC terminals for vesicular glutamate transporter-3 (VGLUT3) or vesicular GABA transporter (VGAT) revealed a significant decrease in CCK-BC synaptic puncta on PN soma in NrCAM-null mice, however no decrease in PV-BC puncta or cell loss. VGLUT3+ CCK-BC puncta were also decreased by Ankyrin B deletion from PNs in Nex1Cre-ERT2:Ank2flox/flox:EGFP mice. A novel CCK-BC reporter mouse expressing tdTomato (tdT) at the Synuclein-γ (Sncg) locus showed NrCAM localized to Sncg + CCK-BCs, and to postsynaptic PN soma in Nex1Cre-ERT2:Ank2+/+:EGFP mice. Results suggest that NrCAM and Ankyrin B contribute to the establishment of connectivity between CCK-BCs and excitatory neurons of the mPFC.
Collapse
Affiliation(s)
- Erik N. Oldre
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Barrett D. Webb
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Justin E. Sperringer
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, CB 7260, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
3
|
Cheheltani S, Islam ST, Malino H, Abera K, Aryal S, Forbes K, Parreno J, Fowler VM. Comparative analysis of rodent lens morphometrics and biomechanical properties. FRONTIERS IN OPHTHALMOLOGY 2025; 5:1562583. [PMID: 40255368 PMCID: PMC12006193 DOI: 10.3389/fopht.2025.1562583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/22/2025]
Abstract
Introduction Proper ocular lens function requires biomechanical flexibility, which is reduced during aging. As increasing lens size has been shown to correlate with lens biomechanical stiffness in aging, we tested the hypothesis that whole lens size determines gross biomechanical stiffness by comparing lenses of varying sizes from three rodent species (mice, rats, and guinea pigs). Methods Coverslip compression assay was performed to measure whole lens biomechanics. Whole mount staining on fixed lenses, followed by confocal microscopy, was conducted to measure lens microstructures. Results Among the three species, guinea pig lenses are the largest, rat lenses are smaller than guinea pig lenses, and mouse lenses are the smallest of the three. We found that rat and guinea pig lenses are stiffer than the much smaller mouse lenses. However, despite guinea pig lenses being larger than rat lenses, whole lens stiffness between guinea pigs and rats is not different. This refutes our hypothesis and indicates that lens size does not solely determine lens stiffness. We next compared lens microstructures, including nuclear size, capsule thickness, epithelial cell area, fiber cell widths, and suture organization between mice, rats, and guinea pigs. The lens nucleus is the largest in guinea pigs, followed by rats, and mice. However, the rat nucleus occupies a larger fraction of the lens. Both lens capsule thickness and fiber cell widths are the largest in guinea pigs, followed by mice and then rats. Epithelial cells are the largest in guinea pigs, and there are no differences between mice and rats. In addition, the lens suture shape appears similar across all three species. Discussion Overall, our data indicates that whole lens size and microstructure morphometrics do not correlate with lens stiffness, indicating that factors contributing to lens biomechanics are complex and likely multifactorial.
Collapse
Affiliation(s)
- Sepideh Cheheltani
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Sadia T. Islam
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Heather Malino
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Kalekidan Abera
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Sandeep Aryal
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Karen Forbes
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE, United States
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| |
Collapse
|
4
|
Panarach C, Netsawang C, Nuchprayoon I, Leecharoenkiat K. Identification and functional analysis of novel SPTB and ANK1 mutations in hereditary spherocytosis patients. Sci Rep 2024; 14:27362. [PMID: 39521890 PMCID: PMC11550412 DOI: 10.1038/s41598-024-78622-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Hereditary spherocytosis (HS) is the most prevalent form of congenital hemolytic anemia, being caused by genetic mutations in genes encoding red blood cell cytoskeletal proteins. Mutations in the ANK1 and SPTB genes are the most common causes of HS.; however, pathogenicity analyses of these mutations remain limited. This study identified three novel heterozygous mutations in 3 HS patients: c.1994 C > A in ANK1, c.5692 C > T, and c.3823delG in SPTB by whole-exome sequencing (WES) and validated by Sanger sequencing. To investigate the functional consequences of these mutations, we studied their pathogenicity using in vitro culture erythroblast derived from CD34 + stem cells. All three mutations lead to the generation of a premature stop codon. Real-time PCR assay revealed that the two SPTB mutations resulted in reduced SPTB mRNA expression, suggesting a potential role for the nonsense-mediated mRNA degradation pathway. For the ANK1 mutation, gene expression was not reduced but was predicted to produce a truncated version of the ANK1 protein. Flow cytometry analysis of red blood cell-derived microparticles (MPs) revealed that HS patients had higher MP levels compared to normal subjects. This study contributes to the current understanding of the molecular mechanisms underlying mutations in the ANK1 and SPTB genes in HS.
Collapse
Affiliation(s)
- Charuwan Panarach
- Programme in Clinical Hematology Sciences, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Chaiwat Netsawang
- Programme in Clinical Hematology Sciences, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Issarang Nuchprayoon
- Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kamonlak Leecharoenkiat
- Oxidation in Red Cell Disorders Research Unit, Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, 154, Rama I Road, Patumwan district, Bangkok, Thailand.
| |
Collapse
|
5
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
6
|
Ghisleni A, Bonilla-Quintana M, Crestani M, Lavagnino Z, Galli C, Rangamani P, Gauthier NC. Mechanically induced topological transition of spectrin regulates its distribution in the mammalian cell cortex. Nat Commun 2024; 15:5711. [PMID: 38977673 PMCID: PMC11231315 DOI: 10.1038/s41467-024-49906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/24/2024] [Indexed: 07/10/2024] Open
Abstract
The cell cortex is a dynamic assembly formed by the plasma membrane and underlying cytoskeleton. As the main determinant of cell shape, the cortex ensures its integrity during passive and active deformations by adapting cytoskeleton topologies through yet poorly understood mechanisms. The spectrin meshwork ensures such adaptation in erythrocytes and neurons by adopting different organizations. Erythrocytes rely on triangular-like lattices of spectrin tetramers, whereas in neurons they are organized in parallel, periodic arrays. Since spectrin is ubiquitously expressed, we exploited Expansion Microscopy to discover that, in fibroblasts, distinct meshwork densities co-exist. Through biophysical measurements and computational modeling, we show that the non-polarized spectrin meshwork, with the intervention of actomyosin, can dynamically transition into polarized clusters fenced by actin stress fibers that resemble periodic arrays as found in neurons. Clusters experience lower mechanical stress and turnover, despite displaying an extension close to the tetramer contour length. Our study sheds light on the adaptive properties of spectrin, which participates in the protection of the cell cortex by varying its densities in response to key mechanical features.
Collapse
Affiliation(s)
- Andrea Ghisleni
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mayte Bonilla-Quintana
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA
| | - Michele Crestani
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Laboratory of Applied Mechanobiology, Department for Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Zeno Lavagnino
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Camilla Galli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Humanitas Cardio Center, IRCCS Humanitas Research Hospital, Rozzano (Milan, Italy
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA.
| | - Nils C Gauthier
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
7
|
Bai SY, Zeng DY, Ouyang M, Zeng Y, Tan W, Xu L. Synaptic cell adhesion molecules contribute to the pathogenesis and progression of fragile X syndrome. Front Cell Neurosci 2024; 18:1393536. [PMID: 39022311 PMCID: PMC11252757 DOI: 10.3389/fncel.2024.1393536] [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: 02/29/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and a monogenic cause of autism spectrum disorders. Deficiencies in the fragile X messenger ribonucleoprotein, encoded by the FMR1 gene, lead to various anatomical and pathophysiological abnormalities and behavioral deficits, such as spine dysmorphogenesis and learning and memory impairments. Synaptic cell adhesion molecules (CAMs) play crucial roles in synapse formation and neural signal transmission by promoting the formation of new synaptic contacts, accurately organizing presynaptic and postsynaptic protein complexes, and ensuring the accuracy of signal transmission. Recent studies have implicated synaptic CAMs such as the immunoglobulin superfamily, N-cadherin, leucine-rich repeat proteins, and neuroligin-1 in the pathogenesis of FXS and found that they contribute to defects in dendritic spines and synaptic plasticity in FXS animal models. This review systematically summarizes the biological associations between nine representative synaptic CAMs and FMRP, as well as the functional consequences of the interaction, to provide new insights into the mechanisms of abnormal synaptic development in FXS.
Collapse
Affiliation(s)
- Shu-Yuan Bai
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - De-Yang Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Ming Ouyang
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Yan Zeng
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Wei Tan
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| | - Lang Xu
- Geriatric Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Alzheimer's Disease, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Murphy KE, Duncan B, Sperringer JE, Zhang E, Haberman V, Wyatt EV, Maness P. Ankyrin B promotes developmental spine regulation in the mouse prefrontal cortex. Cereb Cortex 2023; 33:10634-10648. [PMID: 37642601 PMCID: PMC10560577 DOI: 10.1093/cercor/bhad311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023] Open
Abstract
Postnatal regulation of dendritic spine formation and refinement in cortical pyramidal neurons is critical for excitatory/inhibitory balance in neocortical networks. Recent studies have identified a selective spine pruning mechanism in the mouse prefrontal cortex mediated by class 3 Semaphorins and the L1 cell adhesion molecules, neuron-glia related cell adhesion molecule, Close Homolog of L1, and L1. L1 cell adhesion molecules bind Ankyrin B, an actin-spectrin adaptor encoded by Ankyrin2, a high-confidence gene for autism spectrum disorder. In a new inducible mouse model (Nex1Cre-ERT2: Ank2flox: RCE), Ankyrin2 deletion in early postnatal pyramidal neurons increased spine density on apical dendrites in prefrontal cortex layer 2/3 of homozygous and heterozygous Ankyrin2-deficient mice. In contrast, Ankyrin2 deletion in adulthood had no effect on spine density. Sema3F-induced spine pruning was impaired in cortical neuron cultures from Ankyrin B-null mice and was rescued by re-expression of the 220 kDa Ankyrin B isoform but not 440 kDa Ankyrin B. Ankyrin B bound to neuron-glia related CAM at a cytoplasmic domain motif (FIGQY1231), and mutation to FIGQH inhibited binding, impairing Sema3F-induced spine pruning in neuronal cultures. Identification of a novel function for Ankyrin B in dendritic spine regulation provides insight into cortical circuit development, as well as potential molecular deficiencies in autism spectrum disorder.
Collapse
Affiliation(s)
- Kelsey E Murphy
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Bryce Duncan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Justin E Sperringer
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Erin Zhang
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Victoria Haberman
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Elliott V Wyatt
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| | - Patricia Maness
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Campus Box 7260, Chapel Hill, NC, 27599, United States
| |
Collapse
|
9
|
Pierantozzi E, Raucci L, Buonocore S, Rubino EM, Ding Q, Laurino A, Fiore F, Soldaini M, Chen J, Rossi D, Vangheluwe P, Chen H, Sorrentino V. Skeletal muscle overexpression of sAnk1.5 in transgenic mice does not predispose to type 2 diabetes. Sci Rep 2023; 13:8195. [PMID: 37210436 PMCID: PMC10199891 DOI: 10.1038/s41598-023-35393-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023] Open
Abstract
Genome-wide association studies (GWAS) and cis-expression quantitative trait locus (cis-eQTL) analyses indicated an association of the rs508419 single nucleotide polymorphism (SNP) with type 2 diabetes (T2D). rs508419 is localized in the muscle-specific internal promoter (P2) of the ANK1 gene, which drives the expression of the sAnk1.5 isoform. Functional studies showed that the rs508419 C/C variant results in increased transcriptional activity of the P2 promoter, leading to higher levels of sAnk1.5 mRNA and protein in skeletal muscle biopsies of individuals carrying the C/C genotype. To investigate whether sAnk1.5 overexpression in skeletal muscle might predispose to T2D development, we generated transgenic mice (TgsAnk1.5/+) in which the sAnk1.5 coding sequence was selectively overexpressed in skeletal muscle tissue. TgsAnk1.5/+ mice expressed up to 50% as much sAnk1.5 protein as wild-type (WT) muscles, mirroring the difference reported between individuals with the C/C or T/T genotype at rs508419. However, fasting glucose levels, glucose tolerance, insulin levels and insulin response in TgsAnk1.5/+ mice did not differ from those of age-matched WT mice monitored over a 12-month period. Even when fed a high-fat diet, TgsAnk1.5/+ mice only presented increased caloric intake, but glucose disposal, insulin tolerance and weight gain were comparable to those of WT mice fed a similar diet. Altogether, these data indicate that sAnk1.5 overexpression in skeletal muscle does not predispose mice to T2D susceptibility.
Collapse
Affiliation(s)
- E Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - L Raucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - S Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - E M Rubino
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - Q Ding
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - A Laurino
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - F Fiore
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - M Soldaini
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
| | - J Chen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), 3000, Leuven, Belgium
| | - D Rossi
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100, Siena, Italy
| | - P Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), 3000, Leuven, Belgium
| | - H Chen
- Department of Cardio-Thoracic Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - V Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, 53100, Siena, Italy.
- Interdepartmental Program of Molecular Diagnosis and Pathogenetic Mechanisms of Rare Genetic Diseases, Azienda Ospedaliera Universitaria Senese, 53100, Siena, Italy.
| |
Collapse
|
10
|
Zhou J, Zhang H, Qin Y, Liu T. Severe Microcytic Anemia Caused by Complex Hereditary Spherocytosis and X-Linked Sideroblastic Anemia with Mutations in SPTB and ALAS2 Genes. J Clin Med 2023; 12:jcm12051990. [PMID: 36902777 PMCID: PMC10004689 DOI: 10.3390/jcm12051990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/17/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
We report a case of severe anemia caused by complex hereditary spherocytosis (HS) and X-linked sideroblastic anemia (XLSA) with two mutations in the spectrin beta (SPTB) and 5-aminolevulinic acid synthase (ALAS2) genes. The proband was a 16-year-old male with severe jaundice and microcytic hypochromic anemia since his childhood. He had more severe anemia requiring erythrocyte transfusion, and had no response to vitamin B6 treatment. Next-generation sequencing (NGS) revealed double heterozygous mutations, one in exon 19 (c.3936G > A:p.W1312X) of the SPTB gene and another in exon 2 (c.37A > G:p.K13E) of the ALAS2 gene, and confirmed again by Sanger sequencing. The mutation of ALAS2 (c.37A > G) is inherited from his asymptomatic heterozygous mother, causing amino acid p.K13E, and the mutation has not yet been reported. The mutation of SPTB (c.3936G > A) is a nonsense mutation, leading to a premature termination codon in exon 19, and the mutation in the SPTB gene is not found in any of his relatives, which indicates a de novo monoallelic mutation. Conclusions: The double heterozygous mutations in the SPTB and ALAS2 genes lead to the joint occurrence of HS and XLSA in this patient, and are implicated in the more severe clinical phenotypes.
Collapse
Affiliation(s)
| | | | | | - Ting Liu
- Correspondence: ; Tel.: +86-18980601240
| |
Collapse
|
11
|
Ghisleni A, Bonilla-Quintana M, Crestani M, Fukuzawa A, Rangamani P, Gauthier N. Mechanically induced topological transition of spectrin regulates its distribution in the mammalian cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.02.522381. [PMID: 36712133 PMCID: PMC9881866 DOI: 10.1101/2023.01.02.522381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The cell cortex is a dynamic assembly that ensures cell integrity during passive deformation or active response by adapting cytoskeleton topologies with poorly understood mechanisms. The spectrin meshwork ensures such adaptation in erythrocytes and neurons. Erythrocytes rely on triangular-like lattices of spectrin tetramers, which in neurons are organized in periodic arrays. We exploited Expansion Microscopy to discover that these two distinct topologies can co-exist in other mammalian cells such as fibroblasts. We show through biophysical measurements and computational modeling that spectrin provides coverage of the cortex and, with the intervention of actomyosin, erythroid-like lattices can dynamically transition into condensates resembling neuron-like periodic arrays fenced by actin stress fibers. Spectrin condensates experience lower mechanical stress and turnover despite displaying an extension close to the contour length of the tetramer. Our study sheds light on the adaptive properties of spectrin, which ensures protection of the cortex by undergoing mechanically induced topological transitions.
Collapse
|
12
|
Murphy KE, Zhang EY, Wyatt EV, Sperringer JE, Duncan BW, Maness PF. Doublecortin-Like Kinase 1 Facilitates Dendritic Spine Growth of Pyramidal Neurons in Mouse Prefrontal Cortex. Neuroscience 2023; 508:98-109. [PMID: 36064052 PMCID: PMC10317307 DOI: 10.1016/j.neuroscience.2022.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 01/17/2023]
Abstract
The L1 cell adhesion molecule NrCAM (Neuron-glia related cell adhesion molecule) functions as a co-receptor for secreted class 3 Semaphorins to prune subpopulations of dendritic spines on apical dendrites of pyramidal neurons in the developing mouse neocortex. The developing spine cytoskeleton is enriched in actin filaments, but a small number of microtubules have been shown to enter the spine apparently trafficking vesicles to the membrane. Doublecortin-like kinase 1 (DCLK1) is a member of the Doublecortin (DCX) family of microtubule-binding proteins with serine/threonine kinase activity. To determine if DCLK1 plays a role in spine remodeling, we generated a tamoxifen-inducible mouse line (Nex1Cre-ERT2: DCLK1flox/flox: RCE) to delete microtubule binding isoforms of DCLK1 from pyramidal neurons during postnatal stages of spine development. Homozygous DCLK1 conditional mutant mice exhibited decreased spine density on apical dendrites of pyramidal neurons in the prefrontal cortex (layer 2/3). Mature mushroom spines were selectively decreased upon DCLK1 deletion but dendritic arborization was unaltered. Mutagenesis and binding studies revealed that DCLK1 bound NrCAM at the conserved FIGQY1231 motif in the NrCAM cytoplasmic domain, a known interaction site for the actin-spectrin adaptor Ankyrin. These findings demonstrate in a novel mouse model that DCLK1 facilitates spine growth and maturation on cortical pyramidal neurons in the mouse prefrontal cortex.
Collapse
Affiliation(s)
- Kelsey E Murphy
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States
| | - Erin Y Zhang
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States
| | - Elliott V Wyatt
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States
| | - Justin E Sperringer
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States
| | - Bryce W Duncan
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States
| | - Patricia F Maness
- Department of Biochemistry and Biophysics, and Carolina Institute of Developmental Disabilities, University of North Carolina, School of Medicine at Chapel Hill, United States.
| |
Collapse
|
13
|
Chai Z, Tzingounis AV, Lykotrafitis G. The periodic axon membrane skeleton leads to Na nanodomains but does not impact action potentials. Biophys J 2022; 121:3334-3344. [PMID: 36029000 PMCID: PMC9515372 DOI: 10.1016/j.bpj.2022.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/07/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022] Open
Abstract
Recent work has established that axons have a periodic skeleton structure comprising of azimuthal actin rings connected via longitudinal spectrin tetramer filaments. This structure endows the axon with structural integrity and mechanical stability. Additionally, voltage-gated sodium channels follow the periodicity of the active-spectrin arrangement, spaced ∼190 nm segments apart. The impact of this periodic arrangement of sodium channels on the generation and propagation of action potentials is unknown. To address this question, we simulated an action potential using the Hodgkin-Huxley formalism in a cylindrical compartment, but instead of using a homogeneous distribution of voltage-gated sodium channels in the membrane, we applied the experimentally determined periodic arrangement. We found that the periodic distribution of voltage-gated sodium channels does not significantly affect the generation or propagation of action potentials but instead leads to large, localized sodium action currents caused by high-density sodium nanodomains. Additionally, our simulations show that the distance between periodic sodium channel strips could control axonal excitability, suggesting a previously underappreciated mechanism to regulate neuronal firing properties. Together, this work provides a critical new insight into the role of the periodic arrangement of sodium channels in axons, providing a foundation for future experimental studies.
Collapse
Affiliation(s)
- Zhaojie Chai
- Department of Mechanical Engineering, University of Connecticut, Storrs, Connecticut
| | | | - George Lykotrafitis
- Department of Mechanical Engineering, University of Connecticut, Storrs, Connecticut; Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
14
|
Li D. Role of Spectrin in Endocytosis. Cells 2022; 11:cells11152459. [PMID: 35954302 PMCID: PMC9368487 DOI: 10.3390/cells11152459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Cytoskeletal spectrin is found in (non)erythroid cells. Eukaryotic endocytosis takes place for internalizing cargos from extracellular milieu. The role of spectrin in endocytosis still remains poorly understood. Here, I summarize current knowledge of spectrin function, spectrin-based cytoskeleton and endocytosis of erythrocytes, and highlight how spectrin contributes to endocytosis and working models in different types of cells. From an evolutionary viewpoint, I discuss spectrin and endocytosis in a range of organisms, particularly in plants and yeast where spectrin is absent. Together, the role of spectrin in endocytosis is related to its post-translational modification, movement/rearrangement, elimination (by proteases) and meshwork fencing.
Collapse
Affiliation(s)
- Donghai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
15
|
Munro MJ, Wickremesekera SK, Tan ST, Peng L. Proteomic analysis of low- and high-grade human colon adenocarcinoma tissues and tissue-derived primary cell lines reveals unique biological functions of tumours and new protein biomarker candidates. Clin Proteomics 2022; 19:27. [PMID: 35842572 PMCID: PMC9287856 DOI: 10.1186/s12014-022-09364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background Colon cancer is the third most common cancer and second highest cause of cancer deaths worldwide. The aim of the study was to find new biomarkers for diagnosis, prognosis and therapeutic drug targets for this disease. Methods Four low-grade and four high-grade human colon adenocarcinoma tumours with patient-matched normal colon tissues were analysed. Additionally, tissue-derived primary cell lines were established from each tumour tissue. The cell lines were validated using DNA sequencing to confirm that they are a suitable in vitro model for colon adenocarcinoma based on conserved gene mutations. Label-free quantitation proteomics was performed to compare the proteomes of colon adenocarcinoma samples to normal colon samples, and of colon adenocarcinoma tissues to tissue-derived cell lines to find significantly differentially abundant proteins. The functions enriched within the differentially expressed proteins were assessed using STRING. Proteomics data was validated by Western blotting. Results A total of 4767 proteins were identified across all tissues, and 4711 across primary tissue-derived cell lines. Of these, 3302 proteins were detected in both the tissues and the cell lines. On average, primary cell lines shared about 70% of proteins with their parent tissue, and they retained mutations to key colon adenocarcinoma-related genes and did not diverge far genetically from their parent tissues. Colon adenocarcinoma tissues displayed upregulation of RNA processing, steroid biosynthesis and detoxification, and downregulation of cytoskeletal organisation and loss of normal muscle function. Tissue-derived cell lines exhibited increased interferon-gamma signalling and aberrant ferroptosis. Overall, 318 proteins were significantly up-regulated and 362 proteins significantly down-regulated by comparisons of high-grade with low-grade tumours and low-grade tumour with normal colon tissues from both sample types. Conclusions The differences exhibited between tissues and cell lines highlight the additional information that can be obtained from patient-derived primary cell lines. DNA sequencing and proteomics confirmed that these cell lines can be considered suitable in vitro models of the parent tumours. Various potential biomarkers for colon adenocarcinoma initiation and progression and drug targets were identified and discussed, including seven novel markers: ACSL4, ANK2, AMER3, EXOSC1, EXOSC6, GCLM, and TFRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12014-022-09364-y.
Collapse
Affiliation(s)
- Matthew J Munro
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, 6140, New Zealand.,Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand
| | - Susrutha K Wickremesekera
- Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand.,Upper Gastrointestinal, Hepatobiliary & Pancreatic Section, Department of General Surgery, Wellington Regional Hospital, Wellington, 6021, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Newtown, PO Box 7184, Wellington, 6242, New Zealand. .,Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt, 5040, New Zealand. .,Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3050, Australia.
| | - Lifeng Peng
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, 6140, New Zealand.
| |
Collapse
|
16
|
The spectrin cytoskeleton integrates endothelial mechanoresponses. Nat Cell Biol 2022; 24:1226-1238. [PMID: 35817960 DOI: 10.1038/s41556-022-00953-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Physiological blood flow induces the secretion of vasoactive compounds, notably nitric oxide, and promotes endothelial cell elongation and reorientation parallel to the direction of applied shear. How shear is sensed and relayed to intracellular effectors is incompletely understood. Here, we demonstrate that an apical spectrin network is essential to convey the force imposed by shear to endothelial mechanosensors. By anchoring CD44, spectrins modulate the cell surface density of hyaluronan and sense and translate shear into changes in plasma membrane tension. Spectrins also regulate the stability of apical caveolae, where the mechanosensitive PIEZO1 channels are thought to reside. Accordingly, shear-induced PIEZO1 activation and the associated calcium influx were absent in spectrin-deficient cells. As a result, cell realignment and flow-induced endothelial nitric oxide synthase stimulation were similarly dependent on spectrin. We conclude that the apical spectrin network is not only required for shear sensing but also transmits and distributes the resulting tensile forces to mechanosensors that elicit protective and vasoactive responses.
Collapse
|
17
|
Zou S, Pan BX. Post-synaptic specialization of the neuromuscular junction: junctional folds formation, function, and disorders. Cell Biosci 2022; 12:93. [PMID: 35718785 PMCID: PMC9208267 DOI: 10.1186/s13578-022-00829-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/05/2022] [Indexed: 11/14/2022] Open
Abstract
Post-synaptic specialization is critical to the neurotransmitter release and action potential conduction. The neuromuscular junctions (NMJs) are the synapses between the motor neurons and muscle cells and have a more specialized post-synaptic membrane than synapses in the central nervous system (CNS). The sarcolemma within NMJ folded to form some invagination portions called junctional folds (JFs), and they have important roles in maintaining the post-synaptic membrane structure. The NMJ formation and the acetylcholine receptor (AChR) clustering signal pathway have been extensively studied and reviewed. Although it has been suggested that JFs are related to maintaining the safety factor of neurotransmitter release, the formation mechanism and function of JFs are still unclear. This review will focus on the JFs about evolution, formation, function, and disorders. Anticipate understanding of where they are coming from and where we will study in the future.
Collapse
Affiliation(s)
- Suqi Zou
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China.
- School of Life Sciences, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China.
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China
- School of Life Sciences, Nanchang University, Nanchang, 330031, Jiangxi, P. R. China
| |
Collapse
|
18
|
Bates M, Keller-Findeisen J, Przybylski A, Hüper A, Stephan T, Ilgen P, Cereceda Delgado AR, D'Este E, Egner A, Jakobs S, Sahl SJ, Hell SW. Optimal precision and accuracy in 4Pi-STORM using dynamic spline PSF models. Nat Methods 2022; 19:603-612. [PMID: 35577958 PMCID: PMC9119851 DOI: 10.1038/s41592-022-01465-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022]
Abstract
Coherent fluorescence imaging with two objective lenses (4Pi detection) enables single-molecule localization microscopy with sub-10 nm spatial resolution in three dimensions. Despite its outstanding sensitivity, wider application of this technique has been hindered by complex instrumentation and the challenging nature of the data analysis. Here we report the development of a 4Pi-STORM microscope, which obtains optimal resolution and accuracy by modeling the 4Pi point spread function (PSF) dynamically while also using a simpler optical design. Dynamic spline PSF models incorporate fluctuations in the modulation phase of the experimentally determined PSF, capturing the temporal evolution of the optical system. Our method reaches the theoretical limits for precision and minimizes phase-wrapping artifacts by making full use of the information content of the data. 4Pi-STORM achieves a near-isotropic three-dimensional localization precision of 2–3 nm, and we demonstrate its capabilities by investigating protein and nucleic acid organization in primary neurons and mammalian mitochondria. A dynamic model of the 4Pi point spread function enables localization microscopy with exceptional three-dimensional resolution and a simpler optical design. 4Pi-STORM images of neurons and mitochondria reveal new details of nanoscale protein and nucleic acid organization.
Collapse
Affiliation(s)
- Mark Bates
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany. .,Department of Optical Nanoscopy, Institute for NanoPhotonics, Göttingen, Germany.
| | - Jan Keller-Findeisen
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Adrian Przybylski
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Andreas Hüper
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Till Stephan
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Peter Ilgen
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Angel R Cereceda Delgado
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Elisa D'Este
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Alexander Egner
- Department of Optical Nanoscopy, Institute for NanoPhotonics, Göttingen, Germany
| | - Stefan Jakobs
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Clinic of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Steffen J Sahl
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Stefan W Hell
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany. .,Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg, Germany.
| |
Collapse
|
19
|
Bonacossa-Pereira I, Coakley S, Hilliard MA. Neuron-epidermal attachment protects hyper-fragile axons from mechanical strain. Cell Rep 2022; 38:110501. [PMID: 35263583 DOI: 10.1016/j.celrep.2022.110501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/06/2021] [Accepted: 02/15/2022] [Indexed: 11/03/2022] Open
Abstract
Axons experience significant strain caused by organismal development and movement. A combination of intrinsic mechanical resistance and external shielding by surrounding tissues prevents axonal damage, although the precise mechanisms are unknown. Here, we reveal a neuroprotective function of neuron-epidermal attachment in Caenorhabditis elegans. We show that a gain-of-function mutation in the epidermal hemidesmosome component LET-805/myotactin, in combination with a loss-of-function mutation in UNC-70/β-spectrin, disrupts the uniform attachment and subsequent embedment of sensory axons within the epidermis during development. This generates regions of high tension within axons, leading to spontaneous axonal breaks and degeneration. Completely preventing attachment, by disrupting HIM-4/hemicentin or MEC-5/collagen, eliminates tension and alleviates damage. Finally, we demonstrate that progressive neuron-epidermal attachment via LET-805/myotactin is induced by the axon during development, as well as during regeneration after injury. Together, these results reveal that establishment of uniform neuron-epidermal attachment is critical to protect axons from mechanical strain during development.
Collapse
Affiliation(s)
- Igor Bonacossa-Pereira
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sean Coakley
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Massimo A Hilliard
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
20
|
Mapping the expression of an ANK3 isoform associated with bipolar disorder in the human brain. Transl Psychiatry 2022; 12:45. [PMID: 35091539 PMCID: PMC8799726 DOI: 10.1038/s41398-022-01784-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/09/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
The gene ankyrin-3 (ANK3) has been consistently associated with bipolar disorder (BD) in several genome-wide association studies (GWAS). The exact molecular mechanisms underlying this genetic association remain unknown. The discovery of a loss-of-function variant (rs41283526*G) in an alternatively spliced exon (ENSE00001786716) with a protective effect, suggested that elevated expression of this particular isoform could be a risk factor for developing the disorder. We developed a novel approach for measuring the expression level of all splice forms at a challenging genetic locus using a combination of droplet digital PCR and high-throughput sequencing of indexed PCR amplicons. The combined method was performed on a large collection of 568 postmortem brain samples of BD and SCZ cases and controls. We also studied the expression of the splice forms in a child-development cohort of 41 healthy males. We found that our approach can quantify the splice forms in brain samples, although with less precision than ddPCR. We detected highly significant differences in expression of splice forms and transcription start sites between brain regions, notably with higher expression of the BD-associated isoform in the corpus callosum compared to frontal tissue (mean fold change = 1.80, p < 1e-4). Although the patients in our sample expressed the BD-associated splice form at a similar level to controls, adolescents in our child-development cohort had a clearly higher expression level than younger children (mean fold change = 1.97, p = 5e-3). These results suggest that this ANK3 splice form may play a role in the myelin maturation of the human brain.
Collapse
|
21
|
Pierantozzi E, Szentesi P, Paolini C, Dienes B, Fodor J, Oláh T, Colombini B, Rassier DE, Rubino EM, Lange S, Rossi D, Csernoch L, Bagni MA, Reggiani C, Sorrentino V. Impaired Intracellular Ca 2+ Dynamics, M-Band and Sarcomere Fragility in Skeletal Muscles of Obscurin KO Mice. Int J Mol Sci 2022; 23:1319. [PMID: 35163243 PMCID: PMC8835721 DOI: 10.3390/ijms23031319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Obscurin is a giant sarcomeric protein expressed in striated muscles known to establish several interactions with other proteins of the sarcomere, but also with proteins of the sarcoplasmic reticulum and costameres. Here, we report experiments aiming to better understand the contribution of obscurin to skeletal muscle fibers, starting with a detailed characterization of the diaphragm muscle function, which we previously reported to be the most affected muscle in obscurin (Obscn) KO mice. Twitch and tetanus tension were not significantly different in the diaphragm of WT and Obscn KO mice, while the time to peak (TTP) and half relaxation time (HRT) were prolonged. Differences in force-frequency and force-velocity relationships and an enhanced fatigability are observed in an Obscn KO diaphragm with respect to WT controls. Voltage clamp experiments show that a sarcoplasmic reticulum's Ca2+ release and SERCA reuptake rates were decreased in muscle fibers from Obscn KO mice, suggesting that an impairment in intracellular Ca2+ dynamics could explain the observed differences in the TTP and HRT in the diaphragm. In partial contrast with previous observations, Obscn KO mice show a normal exercise tolerance, but fiber damage, the altered sarcomere ultrastructure and M-band disarray are still observed after intense exercise.
Collapse
Affiliation(s)
- Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Cecilia Paolini
- Department of Neuroscience, Imaging and Clinical Sciences, University Gabriele d’ Annunzio of Chieti, 66100 Chieti, Italy;
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Tamás Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Barbara Colombini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Dilson E. Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC H2W 1S4, Canada;
| | - Egidio Maria Rubino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, La Jolla, CA 92093, USA;
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Maria Angela Bagni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Carlo Reggiani
- Department of Biomedical Science, University of Padova, 35121 Padova, Italy;
- Science and Research Center Koper, Institute for Kinesiology Research, 6000 Koper, Slovenia
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| |
Collapse
|
22
|
Liu J, Liao X, Zhou J, Li B, Xu L, Liu S, Li Y, Yuan D, Hu C, Jiang W, Yan J. A Rare Variant of ANK3 Is Associated With Intracranial Aneurysm. Front Neurol 2021; 12:672570. [PMID: 34248821 PMCID: PMC8267376 DOI: 10.3389/fneur.2021.672570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/01/2021] [Indexed: 12/04/2022] Open
Abstract
Intracranial aneurysm (IA) is a cerebrovascular disorder in which abnormal dilation of a blood vessel results from weakening of the blood vessel wall. The aneurysm may rupture, leading to subarachnoid hemorrhage with severe outcomes. This study was conducted to identify the genetic factors involved in the etiology of IA. Whole-exome sequencing was performed in three IA-aggregate families to identify candidate variants. Further association studies of candidate variants were performed among sporadic cases and controls. Bioinformatic analysis was used to predict the functions of candidate genes and variants. Twenty variants were identified after whole-exome sequencing, among which eight were selected for replicative association studies. ANK3 c.4403G>A (p.R1468H) was significantly associated with IA (odds ratio 4.77; 95% confidence interval 1.94–11.67; p-value = 0.00019). Amino acid R1468 in ANK3 was predicted to be located in the spectrin-binding domain of ankyrin-G and may regulate the migration of vascular endothelial cells and affect cell–cell junctions. Therefore, the variation p.R1468H may cause weakening of the artery walls, thereby accelerating the formation of IA. Thus, ANK3 is a candidate gene highly related to IA.
Collapse
Affiliation(s)
- Junyu Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Liao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jilin Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Bingyang Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Lu Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Songlin Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yifeng Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Dun Yuan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Chongyu Hu
- Department of Neurology, Hunan People's Hospital, Changsha, China
| | - Weixi Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Junxia Yan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| |
Collapse
|
23
|
Ankyrin G organizes membrane components to promote coupling of cell mechanics and glucose uptake. Nat Cell Biol 2021; 23:457-466. [PMID: 33972734 PMCID: PMC8428240 DOI: 10.1038/s41556-021-00677-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/01/2021] [Indexed: 02/03/2023]
Abstract
The response of cells to forces is critical for their function and occurs via rearrangement of the actin cytoskeleton1. Cytoskeletal remodelling is energetically costly2,3, yet how cells signal for nutrient uptake remains undefined. Here we present evidence that force transmission increases glucose uptake by stimulating glucose transporter 1 (GLUT1). GLUT1 recruitment to and retention at sites of force transmission requires non-muscle myosin IIA-mediated contractility and ankyrin G. Ankyrin G forms a bridge between the force-transducing receptors and GLUT1. This bridge is critical for enabling cells under tension to tune glucose uptake to support remodelling of the actin cytoskeleton and formation of an epithelial barrier. Collectively, these data reveal an unexpected mechanism for how cells under tension take up nutrients and provide insight into how defects in glucose transport and mechanics might be linked.
Collapse
|
24
|
Stevens SR, Rasband MN. Ankyrins and neurological disease. Curr Opin Neurobiol 2021; 69:51-57. [PMID: 33485190 DOI: 10.1016/j.conb.2021.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022]
Abstract
Ankyrins are scaffolding proteins widely expressed throughout the nervous system. Ankyrins recruit diverse membrane proteins, including ion channels and cell adhesion molecules, into specialized subcellular membrane domains. These domains are stabilized by ankyrins interacting with the spectrin cytoskeleton. Ankyrin genes are highly associated with a number of neurological disorders, including Alzheimer's disease, schizophrenia, autism spectrum disorders, and bipolar disorder. Here, we discuss ankyrin function and their role in neurological disease. We propose mutations in ankyrins contribute to disease through two primary mechanisms: 1) altered neuronal excitability by disrupting ion channel clustering at key excitable domains, and 2) altered neuronal connectivity via impaired stabilization of membrane proteins.
Collapse
Affiliation(s)
- Sharon R Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
25
|
García-Rodríguez A, Moreno-Olivas F, Marcos R, Tako E, Marques CNH, Mahler GJ. The Role of Metal Oxide Nanoparticles, Escherichia coli, and Lactobacillus rhamnosus on Small Intestinal Enzyme Activity. ENVIRONMENTAL SCIENCE. NANO 2020; 7:3940-3964. [PMID: 33815806 PMCID: PMC8011031 DOI: 10.1039/d0en01001d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Engineered nanomaterials (ENMs) have become common in the food industry, which motivates the need to evaluate ENM effects on human health. Gastrointestinal (GI) in vitro models (e.g. Caco-2, Caco-2/HT29-MTX) have been used in nanotoxicology research. However, the human gut environment is composed of both human cells and the gut microbiota. The goal of this study is to increase the complexity of the Caco-2/HT29-MTX in vitro model by co-culturing human cells with the Gram-positive, commensal Lactobacillus rhamnosus or the Gram-negative, opportunistic Escherichia coli; with the hypothesis that the presence of bacteria would ameliorate the effects of exposure to metal oxide nanoparticles (NPs) such as iron oxide (Fe2O3), silicone dioxide (SiO2), titanium dioxide (TiO2), or zinc oxide (ZnO). To understand this relationship, Caco-2/HT29-MTX cell barriers were acutely co-exposed (4 hours) to bacteria and/or NPs (pristine or in vitro digested). The activity of the brush border membrane (BBM) enzymes intestinal alkaline phosphatase (IAP), aminopeptidase-N (APN), sucrase isomaltase (SI) and the basolateral membrane enzyme (BLM) Na+/K+ ATPase were assessed. Findings show that (i) the human digestion process alters the physicochemical properties of NPs, (ii) large agglomerates of NPs remain entrapped on the apical side of the intestinal barrier, which (iii) affects the activity of BBM enzymes. Interestingly, some NPs effects were attenuated in the presence of either bacterial strains. Confocal microscopy detected bacteria-NPs interactions, which may impede the NP-intestinal cell contact. These results highlight the importance of improving in vitro models to closely mimic the complexities of the human body.
Collapse
Affiliation(s)
- Alba García-Rodríguez
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, 1302, USA
- Department of Genetics and Microbiology, Faculty of Bioscience, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| | - Fabiola Moreno-Olivas
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, 13902, USA
| | - Ricard Marcos
- Department of Genetics and Microbiology, Faculty of Bioscience, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, 08193, Spain
| | - Elad Tako
- Department of Food Science, Cornell University, Stocking Hall, Ithaca, NY, 14853-7201, USA
| | - Cláudia N. H. Marques
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, 1302, USA
| | - Gretchen J. Mahler
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, 13902, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, 13902, USA
| |
Collapse
|
26
|
|
27
|
Complementary mesoscale dynamics of spectrin and acto-myosin shape membrane territories during mechanoresponse. Nat Commun 2020; 11:5108. [PMID: 33037189 PMCID: PMC7547731 DOI: 10.1038/s41467-020-18825-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 09/11/2020] [Indexed: 11/08/2022] Open
Abstract
The spectrin-based membrane skeleton is a major component of the cell cortex. While expressed by all metazoans, its dynamic interactions with the other cortex components, including the plasma membrane or the acto-myosin cytoskeleton, are poorly understood. Here, we investigate how spectrin re-organizes spatially and dynamically under the membrane during changes in cell mechanics. We find spectrin and acto-myosin to be spatially distinct but cooperating during mechanical challenges, such as cell adhesion and contraction, or compression, stretch and osmolarity fluctuations, creating a cohesive cortex supporting the plasma membrane. Actin territories control protrusions and contractile structures while spectrin territories concentrate in retractile zones and low-actin density/inter-contractile regions, acting as a fence that organize membrane trafficking events. We unveil here the existence of a dynamic interplay between acto-myosin and spectrin necessary to support a mesoscale organization of the lipid bilayer into spatially-confined cortical territories during cell mechanoresponse.
Collapse
|
28
|
Abstract
Fodrin and its erythroid cell-specific isoform spectrin are actin-associated fibrous proteins that play crucial roles in the maintenance of structural integrity in mammalian cells, which is necessary for proper cell function. Normal cell morphology is altered in diseases such as various cancers and certain neuronal disorders. Fodrin and spectrin are two-chain (αβ) molecules that are encoded by paralogous genes and share many features but also demonstrate certain differences. Fodrin (in humans, typically a heterodimer of the products of the SPTAN1 and SPTBN1 genes) is expressed in nearly all cell types and is especially abundant in neuronal tissues, whereas spectrin (in humans, a heterodimer of the products of the SPTA1 and SPTB1 genes) is expressed almost exclusively in erythrocytes. To fulfill a role in such a variety of different cell types, it was anticipated that fodrin would need to be a more versatile scaffold than spectrin. Indeed, as summarized here, domains unique to fodrin and its regulation by Ca2+, calmodulin, and a variety of posttranslational modifications (PTMs) endow fodrin with additional specific functions. However, how fodrin structural variations and misregulated PTMs may contribute to the etiology of various cancers and neurodegenerative diseases needs to be further investigated.
Collapse
|
29
|
Li J, Chen K, Zhu R, Zhang M. Structural Basis Underlying Strong Interactions between Ankyrins and Spectrins. J Mol Biol 2020; 432:3838-3850. [DOI: 10.1016/j.jmb.2020.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/18/2020] [Accepted: 04/23/2020] [Indexed: 01/06/2023]
|
30
|
Zhou L, Gao J, Wang H, Shi Y, Xu H, Yan Q, Jing Y, Jiang J, Cai M, Wang H. Correlative dual-color dSTORM/AFM reveals protein clusters at the cytoplasmic side of human bronchial epithelium membranes. NANOSCALE 2020; 12:9950-9957. [PMID: 32356532 DOI: 10.1039/c9nr10931e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The organization of a cell membrane is vital for various functions, such as receptor signaling and membrane traffic. However, the understanding of membrane organization remains insufficient, especially the localizations of specific proteins in the cell membrane. Here, we used correlative super-resolution fluorescence/atomic force microscopy to correlate the distributions of specific proteins Na+/K+-ATPase (NKA, an integral membrane protein) and ankyrin G (AnkG, a scaffolding protein) with the topography of the cytoplasmic side of human bronchial epithelium membranes. Our data showed that NKA and AnkG proteins preferred to localize in the protein islands of membranes. Interestingly, we also found that functional domains composed of specific proteins with a few hundreds of nanometers were formed by assembling protein islands with a few tens of nanometers.
Collapse
Affiliation(s)
- Lulu Zhou
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yang HQ, Pérez-Hernández M, Sanchez-Alonso J, Shevchuk A, Gorelik J, Rothenberg E, Delmar M, Coetzee WA. Ankyrin-G mediates targeting of both Na + and K ATP channels to the rat cardiac intercalated disc. eLife 2020; 9:52373. [PMID: 31934859 PMCID: PMC7299345 DOI: 10.7554/elife.52373] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/11/2020] [Indexed: 12/12/2022] Open
Abstract
We investigated targeting mechanisms of Na+ and KATP channels to the intercalated disk (ICD) of cardiomyocytes. Patch clamp and surface biotinylation data show reciprocal downregulation of each other’s surface density. Mutagenesis of the Kir6.2 ankyrin binding site disrupts this functional coupling. Duplex patch clamping and Angle SICM recordings show that INa and IKATP functionally co-localize at the rat ICD, but not at the lateral membrane. Quantitative STORM imaging show that Na+ and KATP channels are localized close to each other and to AnkG, but not to AnkB, at the ICD. Peptides corresponding to Nav1.5 and Kir6.2 ankyrin binding sites dysregulate targeting of both Na+ and KATP channels to the ICD, but not to lateral membranes. Finally, a clinically relevant gene variant that disrupts KATP channel trafficking also regulates Na+ channel surface expression. The functional coupling between these two channels need to be considered when assessing clinical variants and therapeutics.
Collapse
Affiliation(s)
- Hua-Qian Yang
- Pediatrics, NYU School of Medicine, New York, United States
| | | | - Jose Sanchez-Alonso
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Andriy Shevchuk
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Eli Rothenberg
- Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States
| | - Mario Delmar
- Medicine, NYU School of Medicine, New York, United States.,Cell Biology, NYU School of Medicine, New York, United States
| | - William A Coetzee
- Pediatrics, NYU School of Medicine, New York, United States.,Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, United States.,Neuroscience and Physiology, NYU School of Medicine, New York, United States
| |
Collapse
|
32
|
Mechanistic insights into the interactions of dynein regulator Ndel1 with neuronal ankyrins and implications in polarity maintenance. Proc Natl Acad Sci U S A 2019; 117:1207-1215. [PMID: 31889000 DOI: 10.1073/pnas.1916987117] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ankyrin-G (AnkG), a highly enriched scaffold protein in the axon initial segment (AIS) of neurons, functions to maintain axonal polarity and the integrity of the AIS. At the AIS, AnkG regulates selective intracellular cargo trafficking between soma and axons via interaction with the dynein regulator protein Ndel1, but the molecular mechanism underlying this binding remains elusive. Here we report that Ndel1's C-terminal coiled-coil region (CT-CC) binds to giant neuron-specific insertion regions present in both AnkG and AnkB with 2:1 stoichiometry. The high-resolution crystal structure of AnkB in complex with Ndel1 CT-CC revealed the detailed molecular basis governing the AnkB/Ndel1 complex formation. Mechanistically, AnkB binds with Ndel1 by forming a stable 5-helix bundle dominated by hydrophobic interactions spread across 6 distinct interaction layers. Moreover, we found that AnkG is essential for Ndel1 accumulation at the AIS. Finally, we found that cargo sorting at the AIS can be disrupted by blocking the AnkG/Ndel1 complex formation using a peptide designed based on our structural data. Collectively, the atomic structure of the AnkB/Ndel1 complex together with studies of cargo sorting through the AIS establish the mechanistic basis for AnkG/Ndel1 complex formation and for the maintenance of axonal polarity. Our study will also be valuable for future studies of the interaction between AnkB and Ndel1 perhaps at distal axonal cargo transport.
Collapse
|
33
|
Lattner J, Leng W, Knust E, Brankatschk M, Flores-Benitez D. Crumbs organizes the transport machinery by regulating apical levels of PI(4,5)P 2 in Drosophila. eLife 2019; 8:e50900. [PMID: 31697234 PMCID: PMC6881148 DOI: 10.7554/elife.50900] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
An efficient vectorial intracellular transport machinery depends on a well-established apico-basal polarity and is a prerequisite for the function of secretory epithelia. Despite extensive knowledge on individual trafficking pathways, little is known about the mechanisms coordinating their temporal and spatial regulation. Here, we report that the polarity protein Crumbs is essential for apical plasma membrane phospholipid-homeostasis and efficient apical secretion. Through recruiting βHeavy-Spectrin and MyosinV to the apical membrane, Crumbs maintains the Rab6-, Rab11- and Rab30-dependent trafficking and regulates the lipid phosphatases Pten and Ocrl. Crumbs knock-down results in increased apical levels of PI(4,5)P2 and formation of a novel, Moesin- and PI(4,5)P2-enriched apical membrane sac containing microvilli-like structures. Our results identify Crumbs as an essential hub required to maintain the organization of the apical membrane and the physiological activity of the larval salivary gland.
Collapse
Affiliation(s)
- Johanna Lattner
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Weihua Leng
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Elisabeth Knust
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| | - Marko Brankatschk
- The Biotechnological Center of the TU Dresden (BIOTEC)DresdenGermany
| | - David Flores-Benitez
- Max-Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG)DresdenGermany
| |
Collapse
|
34
|
Lambert MW. Cytoskeletal and nucleoskeletal interacting protein networks play critical roles in cellular function and dysfunction. Exp Biol Med (Maywood) 2019; 244:1233-1239. [PMID: 31657230 DOI: 10.1177/1535370219884875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Muriel W Lambert
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
35
|
Goodman SR, Johnson D, Youngentob SL, Kakhniashvili D. The Spectrinome: The Interactome of a Scaffold Protein Creating Nuclear and Cytoplasmic Connectivity and Function. Exp Biol Med (Maywood) 2019; 244:1273-1302. [PMID: 31483159 DOI: 10.1177/1535370219867269] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We provide a review of Spectrin isoform function in the cytoplasm, the nucleus, the cell surface, and in intracellular signaling. We then discuss the importance of Spectrin’s E2/E3 chimeric ubiquitin conjugating and ligating activity in maintaining cellular homeostasis. Finally we present spectrin isoform subunit specific human diseases. We have created the Spectrinome, from the Human Proteome, Human Reactome and Human Atlas data and demonstrated how it can be a useful tool in visualizing and understanding spectrins myriad of cellular functions.Impact statementSpectrin was for the first 12 years after its discovery thought to be found only in erythrocytes. In 1981, Goodman and colleagues1found that spectrin-like molecules were ubiquitously found in non-erythroid cells leading to a great multitude of publications over the next thirty eight years. The discovery of multiple spectrin isoforms found associated with every cellular compartment, and representing 2-3% of cellular protein, has brought us to today’s understanding that spectrin is a scaffolding protein, with its own E2/E3 chimeric ubiquitin conjugating ligating activity that is involved in virtually every cellular function. We cover the history, localized functions of spectrin isoforms, human diseases caused by mutations, and provide the spectrinome: a useful tool for understanding the myriad of functions for one of the most important proteins in all eukaryotic cells.
Collapse
Affiliation(s)
- Steven R Goodman
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - Daniel Johnson
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - Steven L Youngentob
- Department of Anatomy and Neurobiology, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - David Kakhniashvili
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| |
Collapse
|
36
|
Choi CSW, Souza IA, Sanchez-Arias JC, Zamponi GW, Arbour LT, Swayne LA. Ankyrin B and Ankyrin B variants differentially modulate intracellular and surface Cav2.1 levels. Mol Brain 2019; 12:75. [PMID: 31477143 PMCID: PMC6720858 DOI: 10.1186/s13041-019-0494-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/21/2019] [Indexed: 12/11/2022] Open
Abstract
Ankyrin B (AnkB) is an adaptor and scaffold for motor proteins and various ion channels that is ubiquitously expressed, including in the brain. AnkB has been associated with neurological disorders such as epilepsy and autism spectrum disorder, but understanding of the underlying mechanisms is limited. Cav2.1, the pore-forming subunit of P/Q type voltage gated calcium channels, is a known interactor of AnkB and plays a crucial role in neuronal function. Here we report that wildtype AnkB increased overall Cav2.1 levels without impacting surface Cav2.1 levels in HEK293T cells. An AnkB variant, p.S646F, which we recently discovered to be associated with seizures, further increased overall Cav2.1 levels, again with no impact on surface Cav2.1 levels. AnkB p.Q879R, on the other hand, increased surface Cav2.1 levels in the presence of accessory subunits α2δ1 and β4. Additionally, AnkB p.E1458G decreased surface Cav2.1 irrespective of the presence of accessory subunits. In addition, we found that partial deletion of AnkB in cortex resulted in a decrease in overall Cav2.1 levels, with no change to the levels of Cav2.1 detected in synaptosome fractions. Our work suggests that depending on the particular variant, AnkB regulates intracellular and surface Cav2.1. Notably, expression of the AnkB variant associated with seizure (AnkB p.S646F) caused further increase in intracellular Cav2.1 levels above that of even wildtype AnkB. These novel findings have important implications for understanding the role of AnkB and Cav2.1 in the regulation of neuronal function in health and disease.
Collapse
Affiliation(s)
- Catherine S. W. Choi
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia Canada
| | - Ivana A. Souza
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta Canada
| | - Juan C. Sanchez-Arias
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta Canada
| | - Laura T. Arbour
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia Canada
| | - Leigh Anne Swayne
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia Canada
| |
Collapse
|
37
|
Bose D, Chakrabarti A. Localizing the chaperone activity of erythroid spectrin. Cytoskeleton (Hoboken) 2019; 76:383-397. [DOI: 10.1002/cm.21556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Dipayan Bose
- Crystallography and Molecular Biology DivisionSaha Institute of Nuclear Physics Kolkata India
- Homi Bhabha National Institute Mumbai India
| | - Abhijit Chakrabarti
- Crystallography and Molecular Biology DivisionSaha Institute of Nuclear Physics Kolkata India
- Homi Bhabha National Institute Mumbai India
| |
Collapse
|
38
|
Cordero A, Kanojia D, Miska J, Panek WK, Xiao A, Han Y, Bonamici N, Zhou W, Xiao T, Wu M, Ahmed AU, Lesniak MS. FABP7 is a key metabolic regulator in HER2+ breast cancer brain metastasis. Oncogene 2019; 38:6445-6460. [PMID: 31324889 PMCID: PMC6742563 DOI: 10.1038/s41388-019-0893-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 11/09/2022]
Abstract
Overexpression of human epidermal growth factor receptor 2 (HER2) in breast cancer patients is associated with increased incidence of breast cancer brain metastases (BCBM), but the mechanisms underlying this phenomenon remain unclear. Here, to identify brain-predominant genes critical for the establishment of BCBM, we conducted an in silico screening analysis and identified that increased levels of fatty acid-binding protein 7 (FABP7) correlate with a lower survival and higher incidence of brain metastases in breast cancer patients. We validated these findings using HER2+ BCBM cells compared with parental breast cancer cells. Importantly, through knockdown and overexpression assays, we characterized the role of FABP7 in the BCBM process in vitro and in vivo. Our results uncover a key role of FABP7 in metabolic reprogramming of HER2 + breast cancer cells, supporting a glycolytic phenotype and storage of lipid droplets that enable their adaptation and survival in the brain microenvironment. In addition, FABP7 is shown to be required for upregulation of key metastatic genes and pathways, such as integrins-Src and VEGFA, and for the growth of HER2+ breast cancer cells in the brain microenvironment in vivo. Together, our results support FABP7 as a potential target for the treatment of HER2+ BCBM.
Collapse
Affiliation(s)
- Alex Cordero
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Wojciech K Panek
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Annie Xiao
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yu Han
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicolas Bonamici
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Ting Xiao
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
39
|
Pierantozzi E, Szentesi P, Al-Gaadi D, Oláh T, Dienes B, Sztretye M, Rossi D, Sorrentino V, Csernoch L. Calcium Homeostasis Is Modified in Skeletal Muscle Fibers of Small Ankyrin1 Knockout Mice. Int J Mol Sci 2019; 20:ijms20133361. [PMID: 31323924 PMCID: PMC6651408 DOI: 10.3390/ijms20133361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 11/16/2022] Open
Abstract
Small Ankyrins (sAnk1) are muscle-specific isoforms generated by the Ank1 gene that participate in the organization of the sarcoplasmic reticulum (SR) of striated muscles. Accordingly, the volume of SR tubules localized around the myofibrils is strongly reduced in skeletal muscle fibers of 4- and 10-month-old sAnk1 knockout (KO) mice, while additional structural alterations only develop with aging. To verify whether the lack of sAnk1 also alters intracellular Ca2+ handling, cytosolic Ca2+ levels were analyzed in stimulated skeletal muscle fibers from 4- and 10-month-old sAnk1 KO mice. The SR Ca2+ content was reduced in sAnk1 KO mice regardless of age. The amplitude of the Ca2+ transients induced by depolarizing pulses was decreased in myofibers of sAnk1 KO with respect to wild type (WT) fibers, while their voltage dependence was not affected. Furthermore, analysis of spontaneous Ca2+ release events (sparks) on saponin-permeabilized muscle fibers indicated that the frequency of sparks was significantly lower in fibers from 4-month-old KO mice compared to WT. Furthermore, both the amplitude and spatial spread of sparks were significantly smaller in muscle fibers from both 4- and 10-month-old KO mice compared to WT. These data suggest that the absence of sAnk1 results in an impairment of SR Ca2+ release, likely as a consequence of a decreased Ca2+ store due to the reduction of the SR volume in sAnk1 KO muscle fibers.
Collapse
Affiliation(s)
- Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy
| | - Péter Szentesi
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary
| | - Dána Al-Gaadi
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, H-4002 Debrecen, Hungary
| | - Tamás Oláh
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary
| | - Beatrix Dienes
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary
| | - Mónika Sztretye
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy
| | - László Csernoch
- Department of Physiology, Medical Faculty, University of Debrecen, H-4002 Debrecen, Hungary.
| |
Collapse
|
40
|
Wang Y, Mack JA, Maytin EV. CD44 inhibits α-SMA gene expression via a novel G-actin/MRTF-mediated pathway that intersects with TGFβR/p38MAPK signaling in murine skin fibroblasts. J Biol Chem 2019; 294:12779-12794. [PMID: 31285260 DOI: 10.1074/jbc.ra119.007834] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/25/2019] [Indexed: 01/10/2023] Open
Abstract
Well-regulated differentiation of fibroblasts into myofibroblasts (MF) is critical for skin wound healing. Neoexpression of α-smooth muscle actin (α-SMA), an established marker for MF differentiation, is driven by TGFβ receptor (TGFβR)-mediated signaling. Hyaluronan (HA) and its receptor CD44 may also participate in this process. To further understand this process, primary mouse skin fibroblasts were isolated and treated in vitro with recombinant TGF-β1 (rTGF-β1) to induce α-SMA expression. CD44 expression was also increased. Paradoxically, CD44 knockdown by RNA interference (RNAi) led to increased α-SMA expression and α-SMA-containing stress fibers. Removal of extracellular HA or inhibition of HA synthesis had no effect on α-SMA levels, suggesting a dispensable role for HA. Exploration of mechanisms linking CD44 knockdown to α-SMA induction, using RNAi and chemical inhibitors, revealed a requirement for noncanonical TGFβR signaling through p38MAPK. Decreased monomeric G-actin but increased filamentous F-actin following CD44 RNAi suggested a possible role for myocardin-related transcription factor (MRTF), a known regulator of α-SMA transcription and itself regulated by G-actin binding. CD44 RNAi promoted nuclear accumulation of MRTF and the binding to its transcriptional cofactor SRF. MRTF knockdown abrogated the increased α-SMA expression caused by CD44 RNAi, suggesting that MRTF is required for CD44-mediated regulation of α-SMA. Finally, chemical inhibition of p38MAPK reversed nuclear MRTF accumulation after rTGF-β1 addition or CD44 RNAi, revealing a central involvement of p38MAPK in both cases. We concluded that CD44 regulates α-SMA gene expression through cooperation between two intersecting signaling pathways, one mediated by G-actin/MRTF and the other via TGFβR/p38MAPK.
Collapse
Affiliation(s)
- Yan Wang
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Judith A Mack
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195.,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Edward V Maytin
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195 .,Department of Dermatology, Dermatology and Plastic Surgery Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
41
|
Bapaume L, Laukamm S, Darbon G, Monney C, Meyenhofer F, Feddermann N, Chen M, Reinhardt D. VAPYRIN Marks an Endosomal Trafficking Compartment Involved in Arbuscular Mycorrhizal Symbiosis. FRONTIERS IN PLANT SCIENCE 2019; 10:666. [PMID: 31231402 PMCID: PMC6558636 DOI: 10.3389/fpls.2019.00666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/02/2019] [Indexed: 05/08/2023]
Abstract
Arbuscular mycorrhiza (AM) is a symbiosis between plants and AM fungi that requires the intracellular accommodation of the fungal partner in the host. For reciprocal nutrient exchange, AM fungi form intracellular arbuscules that are surrounded by the peri-arbuscular membrane. This membrane, together with the fungal plasma membrane, and the space in between, constitute the symbiotic interface, over which nutrients are exchanged. Intracellular establishment of AM fungi requires the VAPYRIN protein which is induced in colonized cells, and which localizes to numerous small mobile structures of unknown identity (Vapyrin-bodies). In order to characterize the identity and function of the Vapyrin-bodies we pursued a dual strategy. First, we co-expressed fluorescently tagged VAPYRIN with a range of subcellular marker proteins, and secondly, we employed biochemical tools to identify interacting partner proteins of VAPYRIN. As an important tool for the quantitative analysis of confocal microscopic data sets from co-expression of fluorescent proteins, we developed a semi-automated image analysis pipeline that allows for precise spatio-temporal quantification of protein co-localization and of the dynamics of organelle association from movies. Taken together, these experiments revealed that Vapyrin-bodies have an endosomal identity with trans-Golgi features, and that VAPYRIN interacts with a symbiotic R-SNARE of the VAMP721 family, that localizes to the same compartment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Didier Reinhardt
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
42
|
An expanded proteome of cardiac t-tubules. Cardiovasc Pathol 2019; 42:15-20. [PMID: 31202980 DOI: 10.1016/j.carpath.2019.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/29/2019] [Accepted: 05/17/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Transverse tubules (t-tubules) are important structural elements, derived from sarcolemma, found on all striated myocytes. These specialized organelles create a scaffold for many proteins crucial to the effective propagation of signal in cardiac excitation-contraction coupling. The full protein composition of this region is unknown. METHODS We characterized the t-tubule subproteome using 52,033 immunohistochemical images covering 13,203 proteins from the Human Protein Atlas (HPA) cardiac tissue microarrays. We used HPASubC, a suite of Python tools, to rapidly review and classify each image for a specific t-tubule staining pattern. The tools Gene Cards, String 11, and Gene Ontology Consortium as well as literature searches were used to understand pathways and relationships between the proteins. RESULTS There were 96 likely t-tubule proteins identified by HPASubC. Of these, 12 were matrisome proteins and 3 were mitochondrial proteins. A separate literature search identified 50 known t-tubule proteins. A comparison of the 2 lists revealed only 17 proteins in common, including 8 of the matrisome proteins. String11 revealed that 94 of 127 combined t-tubule proteins generated a single interconnected network. CONCLUSION Using HPASubC and the HPA, we identified 78 novel, putative t-tubule proteins and validated 17 within the literature. This expands and improves our knowledge of this important subcellular structure of the cardiac myocyte. This information can be used to identify new structural targets involved in excitation-contraction coupling that may be altered in disease.
Collapse
|
43
|
Zhang Y, Tzingounis AV, Lykotrafitis G. Modeling of the axon plasma membrane structure and its effects on protein diffusion. PLoS Comput Biol 2019; 15:e1007003. [PMID: 31048841 PMCID: PMC6497228 DOI: 10.1371/journal.pcbi.1007003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/03/2019] [Indexed: 11/30/2022] Open
Abstract
The axon plasma membrane consists of the membrane skeleton, which comprises ring-like actin filaments connected to each other by spectrin tetramers, and the lipid bilayer, which is tethered to the skeleton via, at least, ankyrin. Currently it is unknown whether this unique axon plasma membrane skeleton (APMS) sets the diffusion rules of lipids and proteins in the axon. To answer this question, we developed a coarse-grain molecular dynamics model for the axon that includes the APMS, the phospholipid bilayer, transmembrane proteins (TMPs), and integral monotopic proteins (IMPs) in both the inner and outer lipid layers. We first showed that actin rings limit the longitudinal diffusion of TMPs and the IMPs of the inner leaflet but not of the IMPs of the outer leaflet. To reconcile the experimental observations, which show restricted diffusion of IMPs of the outer leaflet, with our simulations, we conjectured the existence of actin-anchored proteins that form a fence which restricts the longitudinal diffusion of IMPs of the outer leaflet. We also showed that spectrin filaments could modify transverse diffusion of TMPs and IMPs of the inner leaflet, depending on the strength of the association between lipids and spectrin. For instance, in areas where spectrin binds to the lipid bilayer, spectrin filaments would restrict diffusion of proteins within the skeleton corrals. In contrast, in areas where spectrin and lipids are not associated, spectrin modifies the diffusion of TMPs and IMPs of the inner leaflet from normal to confined-hop diffusion. Overall, we showed that diffusion of axon plasma membrane proteins is deeply anisotropic, as longitudinal diffusion is of different type than transverse diffusion. Finally, we investigated how accumulation of TMPs affects diffusion of TMPs and IMPs of both the inner and outer leaflets by changing the density of TMPs. We showed that the APMS structure acts as a fence that restricts the diffusion of TMPs and IMPs of the inner leaflet within the membrane skeleton corrals. Our findings provide insight into how the axon skeleton acts as diffusion barrier and maintains neuronal polarity. The axon plasma membrane skeleton consists of repeated periodic actin ring-like structures along its length connected via spectrin tetramers and anchored to the lipid bilayer at least via ankyrin. However, it is currently unclear whether this structure controls diffusion of lipids and proteins in the axon. Here, we developed a coarse-grain molecular dynamics computational model for the axon plasma membrane that comprises minimal representations for the APMS and the lipid bilayer. In a departure from current models, we found that actin rings limit diffusion of proteins only in the inner membrane leaflet. Then, we showed that actin anchored proteins likely act as “fences” confining diffusion of proteins in the outer leaflet. Our simulations, unexpectedly, also revealed that spectrin filaments could impede transverse diffusion in the inner leaflet of the axon and in some conditions modify diffusion from normal to abnormal. We predicted that diffusion of axon plasma membrane proteins is anisotropic as longitudinal diffusion is of different type than transverse (azimuthal) diffusion. We conclude that the periodic structure of the axon plays a critical role in controlling diffusion of proteins and lipids in the axon plasma membrane.
Collapse
Affiliation(s)
- Yihao Zhang
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, United States of America
| | - Anastasios V. Tzingounis
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States of America
| | - George Lykotrafitis
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, United States of America
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States of America
- * E-mail:
| |
Collapse
|
44
|
Grimes KM, Prasad V, McNamara JW. Supporting the heart: Functions of the cardiomyocyte's non-sarcomeric cytoskeleton. J Mol Cell Cardiol 2019; 131:187-196. [PMID: 30978342 DOI: 10.1016/j.yjmcc.2019.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
The non-contractile cytoskeleton in cardiomyocytes is comprised of cytoplasmic actin, microtubules, and intermediate filaments. In addition to providing mechanical support to these cells, these structures are important effectors of tension-sensing and signal transduction and also provide networks for the transport of proteins and organelles. The majority of our knowledge on the function and structure of these cytoskeletal networks comes from research on proliferative cell types. However, in recent years, researchers have begun to show that there are important cardiomyocyte-specific functions of the cytoskeleton. Here we will discuss the current state of cytoskeletal biology in cardiomyocytes, as well as research from other cell types, that together suggest there is a wealth of knowledge on cardiac health and disease waiting to be uncovered through exploration of the complex signaling networks of cardiomyocyte non-sarcomeric cytoskeletal proteins.
Collapse
Affiliation(s)
- Kelly M Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - James W McNamara
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
45
|
The cell-cell junctions of mammalian testes: II. The lamellar smooth muscle monolayer cells of the peritubular wall are laterally connected by vertical adherens junctions-a novel architectonic cell-cell junction system. Cell Tissue Res 2018; 375:451-482. [PMID: 30591979 DOI: 10.1007/s00441-018-2968-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022]
Abstract
The testes of sexually mature males of six mammalian species (men, bulls, boars, rats, mice, guinea pigs) have been studied using biochemical as well as light and electron microscopical techniques, in particular immunolocalizations. In these tissues, the peritubular walls represent lamellar encasement structures wrapped around the seminiferous tubules as a bandage system of extracellular matrix layers, alternating with monolayers of very flat polyhedral "lamellar smooth muscle cells" (LSMCs), the number of which varies in different species from 1 to 5 or 6. These LSMCs are complete SMCs containing smooth muscle α-actin (SMA), myosin light and heavy chains, α-actinin, tropomyosin, smoothelin, intermediate-sized filament proteins desmin and/or vimentin, filamin, talin, dystrophin, caldesmon, calponin, and protein SM22α, often also cytokeratins 8 and 18. In the monolayers, the LSMCs are connected by adherens junctions (AJs) based on cadherin-11, in some species also with P-cadherin and/or E-cadherin, which are anchored in cytoplasmic plaques containing β-catenin and other armadillo proteins, in some species also striatin family proteins, protein myozap and/or LUMA. The LSMC cytoplasm is rich in myofilament bundles, which in many regions are packed in paracrystalline arrays, as well as in "dense bodies," "focal adhesions," and caveolae. In addition to some AJ-like end-on-end contacts, the LSMCs are laterally connected by numerous vertical AJ-like junctions located in variously sized and variously shaped, overlapping (alter super alterum) lamelliform cell protrusions. Consequently, the LSMCs of the peritubular wall monolayers are SMCs sensu stricto which are laterally connected by a novel architectonic system of arrays of vertical AJs located in overlapping cell protrusions.
Collapse
|
46
|
Ankyrin-G regulated epithelial phenotype is required for mouse lens morphogenesis and growth. Dev Biol 2018; 446:119-131. [PMID: 30562487 DOI: 10.1016/j.ydbio.2018.12.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 11/24/2022]
Abstract
Epithelial cell polarity, adhesion, proliferation, differentiation and survival are essential for morphogenesis of various organs and tissues including the ocular lens. The molecular mechanisms regulating the lens epithelial phenotype however, are not well understood. Here we investigated the role of scaffolding protein ankyrin-G (AnkG) in mouse lens development by conditional suppression of AnkG expression using the Cre-LoxP recombination approach. AnkG, which serves to link integral membrane proteins to the spectrin/actin cytoskeleton, was found to distribute predominantly to the lateral membranes of lens epithelium with several isoforms of the protein being detected in the mouse lens. Conditional deficiency of AnkG impaired mouse lens morphogenesis starting from embryonic stage E15.5, with neonatal (P1) AnkG cKO lenses exhibiting overt abnormalities in shape, size, epithelial cell height, sheet length and lateral membrane assembly together with defective fiber cell orientation relative to lenses from littermate AnkG floxed or Cre expressing mice. Severe disruptions in E-cadherin/β-catenin-based adherens junctions, and the membrane organization of spectrin-actin cytoskeleton, ZO-1, connexin-50 and Na+-K+-ATPase were noted in AnkG deficient lenses, along with detection in lens epithelium of α-smooth muscle actin, a marker of epithelial to mesenchymal transition. Moreover, lens epithelial cell proliferation and survival were severely compromised while differentiation appears to be normal in AnkG deficient mouse lenses. Collectively, these results indicate that AnkG regulates establishment of the epithelial phenotype via lateral membrane assembly, stabilization of E-cadherin-based cell-cell junctions, polarity and membrane organization of transport and adhesion proteins and the spectrin-actin skeleton, and provide evidence for an obligatory role for AnkG in lens morphogenesis and growth.
Collapse
|
47
|
Parreno J, Fowler VM. Multifunctional roles of tropomodulin-3 in regulating actin dynamics. Biophys Rev 2018; 10:1605-1615. [PMID: 30430457 DOI: 10.1007/s12551-018-0481-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022] Open
Abstract
Tropomodulins (Tmods) are proteins that cap the slow-growing (pointed) ends of actin filaments (F-actin). The basis for our current understanding of Tmod function comes from studies in cells with relatively stable and highly organized F-actin networks, leading to the view that Tmod capping functions principally to preserve F-actin stability. However, not only is Tmod capping dynamic, but it also can play major roles in regulating diverse cellular processes involving F-actin remodeling. Here, we highlight the multifunctional roles of Tmod with a focus on Tmod3. Like other Tmods, Tmod3 binds tropomyosin (Tpm) and actin, capping pure F-actin at submicromolar and Tpm-coated F-actin at nanomolar concentrations. Unlike other Tmods, Tmod3 can also bind actin monomers and its ability to bind actin is inhibited by phosphorylation of Tmod3 by Akt2. Tmod3 is ubiquitously expressed and is present in a diverse array of cytoskeletal structures, including contractile structures such as sarcomere-like units of actomyosin stress fibers and in the F-actin network encompassing adherens junctions. Tmod3 participates in F-actin network remodeling in lamellipodia during cell migration and in the assembly of specialized F-actin networks during exocytosis. Furthermore, Tmod3 is required for development, regulating F-actin mesh formation during meiosis I of mouse oocytes, erythroblast enucleation in definitive erythropoiesis, and megakaryocyte morphogenesis in the mouse fetal liver. Thus, Tmod3 plays vital roles in dynamic and stable F-actin networks in cell physiology and development, with further research required to delineate the mechanistic details of Tmod3 regulation in the aforementioned processes, or in other yet to be discovered processes.
Collapse
Affiliation(s)
- Justin Parreno
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Velia M Fowler
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
48
|
Calpain inhibition ameliorates scald burn-induced acute lung injury in rats. BURNS & TRAUMA 2018; 6:28. [PMID: 30338266 PMCID: PMC6174571 DOI: 10.1186/s41038-018-0130-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022]
Abstract
Background The molecular pattern of severe burn-induced acute lung injury, characterized by cell structure damage and leukocyte infiltration, remains unknown. This study aimed to determine whether calpain, a protease involved in both processes, mediates severe burn-induced acute lung injury. Methods Rats received full-thickness scald burns covering 30% of the total body surface area, followed by instant fluid resuscitation. MDL28170 (Tocris Bioscience), an inhibitor of calpain, was given intravenously 1 h before or after the scald burn. The histological score, wet/dry weight ratio, and caspase-3 activity were examined to evaluate the degree of lung damage. Calpain activity and its source were detected by an assay kit and immunofluorescence staining. The proteolysis of membrane skeleton proteins α-fodrin and ankyrin-B, which are substrates of calpain, was measured by Western blot. Results Time-course studies showed that tissue damage reached a peak between 1 and 6 h post-scald burn and gradually diminished at 24 h. More importantly, calpain activity reached peak levels at 1 h and was maintained until 24 h, paralleled by lung damage to some extent. Western blot showed that the levels of the proteolyzed forms of α-fodrin and ankyrin-B correlated well with the degree of damage. MDL28170 at a dose of 3 mg/kg b. w. given 1 h before burn injury not only antagonized the increase in calpain activity but also ameliorated scald burn-induced lung injury, including the degradation of α-fodrin and ankyrin-B. Immunofluorescence images revealed calpain 1 and CD45 double-positive cells in the lung tissue of rats exposed to scald burn injury, suggesting that leukocytes were a dominant source of calpain. Furthermore, this change was blocked by MDL28170. Finally, MDL28170 given at 1 h post-scald burn injury significantly ameliorated the wet/dry weight ratio compared with burn injury alone. Conclusions Calpain, a product of infiltrating leukocytes, is a mediator of scald burn-induced acute lung injury that involves enhancement of inflammation and proteolysis of membrane skeleton proteins. Its late effects warrant further study.
Collapse
|
49
|
Lechuga S, Amin PH, Wolen AR, Ivanov AI. Adducins inhibit lung cancer cell migration through mechanisms involving regulation of cell-matrix adhesion and cadherin-11 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:395-408. [PMID: 30290240 DOI: 10.1016/j.bbamcr.2018.10.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 09/16/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
Cell migration is a critical mechanism controlling tissue morphogenesis, epithelial wound healing and tumor metastasis. Migrating cells depend on orchestrated remodeling of the plasma membrane and the underlying actin cytoskeleton, which is regulated by the spectrin-adducin-based membrane skeleton. Expression of adducins is altered during tumorigenesis, however, their involvement in metastatic dissemination of tumor cells remains poorly characterized. This study investigated the roles of α-adducin (ADD1) and γ-adducin (ADD3) in regulating migration and invasion of non-small cell lung cancer (NSCLC) cells. ADD1 was mislocalized, whereas ADD3 was markedly downregulated in NSCLC cells with the invasive mesenchymal phenotype. CRISPR/Cas9-mediated knockout of ADD1 and ADD3 in epithelial-type NSCLC and normal bronchial epithelial cells promoted their Boyden chamber migration and Matrigel invasion. Furthermore, overexpression of ADD1, but not ADD3, in mesenchymal-type NSCLC cells decreased cell migration and invasion. ADD1-overexpressing NSCLC cells demonstrated increased adhesion to the extracellular matrix (ECM), accompanied by enhanced assembly of focal adhesions and hyperphosphorylation of Src and paxillin. The increased adhesiveness and decreased motility of ADD1-overexpressing cells were reversed by siRNA-mediated knockdown of Src. By contrast, the accelerated migration of ADD1 and ADD3-depleted NSCLC cells was ECM adhesion-independent and was driven by the upregulated expression of pro-motile cadherin-11. Overall, our findings reveal a novel function of adducins as negative regulators of NSCLC cell migration and invasion, which could be essential for limiting lung cancer progression and metastasis.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH 44195, United States of America; Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Parth H Amin
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Aaron R Wolen
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute of Cleveland Clinic Foundation, Cleveland, OH 44195, United States of America; Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| |
Collapse
|
50
|
Duan R, Kim JH, Shilagardi K, Schiffhauer ES, Lee DM, Son S, Li S, Thomas C, Luo T, Fletcher DA, Robinson DN, Chen EH. Spectrin is a mechanoresponsive protein shaping fusogenic synapse architecture during myoblast fusion. Nat Cell Biol 2018; 20:688-698. [PMID: 29802406 PMCID: PMC6397639 DOI: 10.1038/s41556-018-0106-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 04/18/2018] [Indexed: 12/24/2022]
Abstract
Spectrin is a membrane skeletal protein best known for its structural role in maintaining cell shape and protecting cells from mechanical damage. Here, we report that α/βH-spectrin (βH is also called karst) dynamically accumulates and dissolves at the fusogenic synapse between fusing Drosophila muscle cells, where an attacking fusion partner invades its receiving partner with actin-propelled protrusions to promote cell fusion. Using genetics, cell biology, biophysics and mathematical modelling, we demonstrate that spectrin exhibits a mechanosensitive accumulation in response to shear deformation, which is highly elevated at the fusogenic synapse. The transiently accumulated spectrin network functions as a cellular fence to restrict the diffusion of cell-adhesion molecules and a cellular sieve to constrict the invasive protrusions, thereby increasing the mechanical tension of the fusogenic synapse to promote cell membrane fusion. Our study reveals a function of spectrin as a mechanoresponsive protein and has general implications for understanding spectrin function in dynamic cellular processes.
Collapse
Affiliation(s)
- Rui Duan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Regenerative Medicine in Sports Science, School of Sports Science, South China Normal University, Guangzhou, China
| | - Ji Hoon Kim
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Khurts Shilagardi
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric S Schiffhauer
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donghoon M Lee
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Shuo Li
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Claire Thomas
- Departments of Biology and of Biochemistry and Molecular Biology, Penn State University, University Park, PA, USA
| | - Tianzhi Luo
- Department of Modern Mechanics, University of Science and Technology of China, Hefei, China
| | - Daniel A Fletcher
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth H Chen
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|