Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
Add more filters

Publication year range
1.
Cell Mol Biol Lett ; 25: 3, 2020.
Article in English | MEDLINE | ID: mdl-32042281

ABSTRACT

BACKGROUND: Precise coordination of cytoskeletal components and dynamic control of cell adhesion and migration are required for crucial cell processes such as differentiation and morphogenesis. We investigated the potential involvement of αII-spectrin, a ubiquitous scaffolding element of the membrane skeleton, in the adhesion and angiogenesis mechanism. METHODS: The cell models were primary human umbilical vein endothelial cells (HUVECs) and a human dermal microvascular endothelial cell line (HMEC-1). After siRNA- and shRNA-mediated knockdown of αII-spectrin, we assessed its expression and that of its partners and adhesion proteins using western blotting. The phenotypes of the control and spectrin-depleted cells were examined using immunofluorescence and video microscopy. Capillary tube formation was assessed using the thick gel Matrigel matrix-based method and a microscope equipped with a thermostatic chamber and a Nikon Biostation System camera. RESULTS: Knockdown of αII-spectrin leads to: modified cell shape; actin cytoskeleton organization with the presence of peripheral actin patches; and decreased formation of stress fibers. Spectrin deficiency affects cell adhesion on laminin and fibronectin and cell motility. This included modification of the localization of adhesion molecules, such as αVƟ3- and α5-integrins, and organization of adhesion structures, such as focal points. Deficiency of αII-spectrin can also affect the complex mechanism of in vitro capillary tube formation, as demonstrated in a model of angiogenesis. Live imaging revealed that impairment of capillary tube assembly was mainly associated with a significant decrease in cell projection length and stability. αII-spectrin depletion is also associated with significantly decreased expression of three proteins involved in capillary tube formation and assembly: VE-cadherin, MCAM and Ɵ3-integrin. CONCLUSION: Our data confirm the role of αII-spectrin in the control of cell adhesion and spreading. Moreover, our findings further support the participation of αII-spectrin in capillary tube formation in vitro through control of adhesion molecules, such as integrins. This indicates a new function of αII-spectrin in angiogenesis.


Subject(s)
Actin Cytoskeleton/metabolism , Capillaries/metabolism , Cell Adhesion/physiology , Endothelial Cells/metabolism , Neovascularization, Physiologic , Spectrin/metabolism , Antigens, CD/metabolism , CD146 Antigen/metabolism , Cadherins/metabolism , Capillaries/growth & development , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cell Shape , Endothelial Cells/cytology , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha Chains/metabolism , Integrin beta3/metabolism , Laminin/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Neovascularization, Physiologic/genetics , RNA, Small Interfering , Spectrin/deficiency , Spectrin/genetics , Stress Fibers/metabolism
2.
Hum Genet ; 136(7): 903-910, 2017 07.
Article in English | MEDLINE | ID: mdl-28540413

ABSTRACT

Congenital myopathies are a heterogeneous group of muscle disorders that are often genetically determined. Here, we investigated a boy with congenital myopathy, deafness, and neuropathy from a consanguineous Kurdish family by autozygosity mapping and whole exome sequencing. We found a homozygous nonsense mutation in SPTBN4 [c.1597C>T, NM_020971.2; p.(Q533*), NP_066022.2; ClinVar SUB2292235] encoding ƟIV-spectrin, a non-erythrocytic member of the Ɵ-spectrin family. Western blot confirmed the absence of the full-length 288Ā kDa isoform in muscle and of a specific 72Ā kDa isoform in fibroblasts. Clinical symptoms of the patient largely corresponded to those described for the quivering mouse, a loss-of-function animal model. Since the human phenotype of ƟIV-spectrin deficiency included a myopathy with incomplete congenital fiber-type disproportion, we investigated muscle of the quivering (qv4J) mouse and found complete absence of type 1 fibers (fiber-type 2 uniformity). Immunohistology confirmed expression of ƟIV-spectrin in normal human and mouse muscle at the sarcolemma and its absence in patient and quivering (qv4J) mouse. SPTBN4 mRNA-expression levels in healthy skeletal muscle were found in the range of other regulatory proteins. More patients have to be described to confirm the triad of congenital myopathy, neuropathy and deafness as the defining symptom complex for ƟIV-spectrin deficiency.


Subject(s)
Congenital Abnormalities/genetics , Deafness/genetics , Genes, Recessive , Muscular Diseases/genetics , Nerve Tissue Proteins/genetics , Spectrin/genetics , Amino Acid Sequence , Animals , Chromosome Mapping , Codon, Nonsense , Gene Expression Profiling , Genome, Human , Humans , Male , Mice , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Pedigree , Sequence Analysis, DNA , Spectrin/deficiency , Spectrin/metabolism
3.
J Cell Sci ; 125(Pt 4): 919-31, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22275434

ABSTRACT

Although columnar epithelial cells are known to acquire an elongated shape, the mechanisms involved in this morphological feature have not yet been completely elucidated. Using columnar human intestinal Caco2 cells, it was established here that the levels of drebrin E, an actin-binding protein, increase in the terminal web both in vitro and in vivo during the formation of the apical domain. Drebrin E depletion was found to impair cell compaction and elongation processes in the monolayer without affecting cell polarity or the formation of tight junctions. Decreasing the drebrin E levels disrupted the normal subapical F-actin-myosin-IIB-ƟII-spectrin network and the apical accumulation of EB3, a microtubule-plus-end-binding protein. Decreasing the EB3 levels resulted in a similar elongation phenotype to that resulting from depletion of drebrin E, without affecting cell compaction processes or the pattern of distribution of F-actin-myosin-IIB. In addition, EB3, myosin IIB and ƟII spectrin were found to form a drebrin-E-dependent complex. Taken together, these data suggest that this complex connects the F-actin and microtubule networks apically during epithelial cell morphogenesis, while drebrin E also contributes to stabilizing the actin-based terminal web.


Subject(s)
Cell Shape/physiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Neuropeptides/metabolism , Spectrin/metabolism , Actins/metabolism , Caco-2 Cells , Cell Polarity/physiology , Humans , Microtubule-Associated Proteins , Neuropeptides/deficiency , Nonmuscle Myosin Type IIB/metabolism , Spectrin/deficiency , Tight Junctions
4.
Proc Natl Acad Sci U S A ; 108(19): 8009-14, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518878

ABSTRACT

During peripheral nerve development, Schwann cells ensheathe axons and form myelin to enable rapid and efficient action potential propagation. Although myelination requires profound changes in Schwann cell shape, how neuron-glia interactions converge on the Schwann cell cytoskeleton to induce these changes is unknown. Here, we demonstrate that the submembranous cytoskeletal proteins αII and ƟII spectrin are polarized in Schwann cells and colocalize with signaling molecules known to modulate myelination in vitro. Silencing expression of these spectrins inhibited myelination in vitro, and remyelination in vivo. Furthermore, myelination was disrupted in motor nerves of zebrafish lacking αII spectrin. Finally, we demonstrate that loss of spectrin significantly reduces both F-actin in the Schwann cell cytoskeleton and the Nectin-like protein, Necl4, at the contact site between Schwann cells and axons. Therefore, we propose αII and ƟII spectrin in Schwann cells integrate the neuron-glia interactions mediated by membrane proteins into the actin-dependent cytoskeletal rearrangements necessary for myelination.


Subject(s)
Myelin Sheath/physiology , Peripheral Nerves/physiology , Schwann Cells/physiology , Spectrin/physiology , Actins/antagonists & inhibitors , Actins/physiology , Animals , Base Sequence , Cell Polarity , Cytoskeleton/physiology , Gene Knockdown Techniques , Mutation , RNA Interference , Rats , Rats, Sprague-Dawley , Schwann Cells/cytology , Sciatic Nerve/cytology , Sciatic Nerve/injuries , Sciatic Nerve/physiology , Spectrin/antagonists & inhibitors , Spectrin/deficiency , Spectrin/genetics , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
5.
Proc Natl Acad Sci U S A ; 107(13): 6022-7, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20231455

ABSTRACT

The spectrin membrane skeleton controls the disposition of selected membrane channels, receptors, and transporters. In the brain betaIII spectrin binds directly to the excitatory amino acid transporter (EAAT4), the glutamate receptor delta, and other proteins. Mutations in betaIII spectrin link strongly to human spinocerebellar ataxia type 5 (SCA5), correlating with alterations in EAAT4. We have explored the mechanistic basis of this phenotype by targeted gene disruption of Spnb3. Mice lacking intact betaIII spectrin develop normally. By 6 months they display a mild nonprogressive ataxia. By 1 year most Spnb3(-/-) animals develop a myoclonic seizure disorder with significant reductions of EAAT4, EAAT1, GluRdelta, IP3R, and NCAM140. Other synaptic proteins are normal. The cerebellum displays increased dark Purkinje cells (PC), a thin molecular layer, fewer synapses, a loss of dendritic spines, and a 2-fold expansion of the PC dendrite diameter. Membrane and expanded Golgi profiles fill the PC dendrite and soma, and both regions accumulate EAAT4. Correlating with the seizure disorder are enhanced hippocampal levels of neuropeptide Y and EAAT3 and increased calpain proteolysis of alphaII spectrin. It appears that betaIII spectrin disruption impairs synaptogenesis by disturbing the intracellular pathways selectively regulating protein trafficking to the synapse. The mislocalization of these proteins secondarily disrupts glutamate transport dynamics, leading to seizures, neuronal damage, and compensatory changes in EAAT3 and neuropeptide Y.


Subject(s)
Ataxia/etiology , Seizures/etiology , Spectrin/deficiency , Animals , Ataxia/genetics , Ataxia/physiopathology , Base Sequence , Brain/metabolism , Brain/physiopathology , Brain/ultrastructure , DNA Primers/genetics , Disease Models, Animal , Excitatory Amino Acid Transporter 4/metabolism , Female , Gene Targeting , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Phenotype , Seizures/genetics , Seizures/physiopathology , Spectrin/genetics , Spectrin/physiology , Spinocerebellar Ataxias/etiology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Synapses/physiology , Synapses/ultrastructure
6.
J Neurosci ; 31(46): 16581-90, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090485

ABSTRACT

Mutations in the gene encoding Ɵ-III spectrin give rise to spinocerebellar ataxia type 5, a neurodegenerative disease characterized by progressive thinning of the molecular layer, loss of Purkinje cells and increasing motor deficits. A mouse lacking full-length Ɵ-III spectrin (Ɵ-IIIĆ¢ĀĀ»/Ć¢ĀĀ») displays a similar phenotype. In vitro and in vivo analyses of Purkinje cells lacking Ɵ-III spectrin, reveal a critical role for Ɵ-III spectrin in Purkinje cell morphological development. Disruption of the normally well ordered dendritic arborization occurs in Purkinje cells from Ɵ-IIIĆ¢ĀĀ»/Ć¢ĀĀ» mice, specifically showing a loss of monoplanar organization, smaller average dendritic diameter and reduced densities of Purkinje cell spines and synapses. Early morphological defects appear to affect distribution of dendritic, but not axonal, proteins. This study confirms that thinning of the molecular layer associated with disease pathogenesis is a consequence of Purkinje cell dendritic degeneration, as Purkinje cells from 8-month-old Ɵ-IIIĆ¢ĀĀ»/Ć¢ĀĀ» mice have drastically reduced dendritic volumes, surface areas and total dendritic lengths compared with 5- to 6-week-old Ɵ-IIIĆ¢ĀĀ»/Ć¢ĀĀ» mice. These findings highlight a critical role of Ɵ-III spectrin in dendritic biology and are consistent with an early developmental defect in Ɵ-IIIĆ¢ĀĀ»/Ć¢ĀĀ» mice, with abnormal Purkinje cell dendritic morphology potentially underlying disease pathogenesis.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dendrites/ultrastructure , Dendritic Spines/metabolism , Purkinje Cells/cytology , Spectrin/metabolism , Age Factors , Animals , Animals, Newborn , Calbindins , Excitatory Amino Acid Transporter 4/metabolism , Gene Expression Regulation, Developmental/genetics , Glucose Transporter Type 2/metabolism , In Vitro Techniques , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Patch-Clamp Techniques , Phosphate Transport Proteins/metabolism , S100 Calcium Binding Protein G/metabolism , Silver Staining/methods , Sodium Channels/metabolism , Spectrin/deficiency , Vesicular Glutamate Transport Protein 1/metabolism
7.
Blood ; 115(9): 1804-14, 2010 Mar 04.
Article in English | MEDLINE | ID: mdl-20056793

ABSTRACT

Five spontaneous, allelic mutations in the alpha-spectrin gene, Spna1, have been identified in mice (spherocytosis [sph], sph(1J), sph(2J), sph(2BC), sph(Dem)). All cause severe hemolytic anemia. Here, analysis of 3 new alleles reveals previously unknown consequences of red blood cell (RBC) spectrin deficiency. In sph(3J), a missense mutation (H2012Y) in repeat 19 introduces a cryptic splice site resulting in premature termination of translation. In sph(Ihj), a premature stop codon occurs (Q1853Stop) in repeat 18. Both mutations result in markedly reduced RBC membrane spectrin content, decreased band 3, and absent beta-adducin. Reevaluation of available, previously described sph alleles reveals band 3 and adducin deficiency as well. In sph(4J), a missense mutation occurs in the C-terminal EF hand domain (C2384Y). Notably, an equally severe hemolytic anemia occurs despite minimally decreased membrane spectrin with normal band 3 levels and present, although reduced, beta-adducin. The severity of anemia in sph(4J) indicates that the highly conserved cysteine residue at the C-terminus of alpha-spectrin participates in interactions critical to membrane stability. The data reinforce the notion that a membrane bridge in addition to the classic protein 4.1-p55-glycophorin C linkage exists at the RBC junctional complex that involves interactions between spectrin, adducin, and band 3.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/deficiency , Anion Exchange Protein 1, Erythrocyte/genetics , Calmodulin-Binding Proteins/deficiency , Calmodulin-Binding Proteins/genetics , Mutation , Spectrin/deficiency , Spectrin/genetics , Spherocytosis, Hereditary/blood , Spherocytosis, Hereditary/genetics , Alleles , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Calmodulin-Binding Proteins/blood , Codon, Nonsense , DNA Primers/genetics , Erythrocyte Membrane/metabolism , Erythrocytes, Abnormal/metabolism , Erythrocytes, Abnormal/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Mutant Strains , Molecular Sequence Data , Mutation, Missense , RNA Stability/genetics , Sequence Homology, Amino Acid , Spherocytosis, Hereditary/pathology
8.
J Neurosci ; 30(14): 4857-67, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371805

ABSTRACT

Mutations in SPTBN2, the gene encoding beta-III spectrin, cause spinocerebellar ataxia type 5 in humans (SCA5), a neurodegenerative disorder resulting in loss of motor coordination. How these mutations give rise to progressive ataxia and what the precise role beta-III spectrin plays in normal cerebellar physiology are unknown. We developed a mouse lacking full-length beta-III spectrin and found that homozygous mice reproduced features of SCA5 including gait abnormalities, tremor, deteriorating motor coordination, Purkinje cell loss, and cerebellar atrophy (molecular layer thinning). In vivo analysis reveals an age-related reduction in simple spike firing rate in surviving beta-III(-/-) Purkinje cells, whereas in vitro studies show these neurons to have reduced spontaneous firing, smaller sodium currents, and dysregulation of glutamatergic neurotransmission. Our data suggest an early loss of EAAT4- (protein interactor of beta-III spectrin) and a subsequent loss of GLAST-mediated uptake may play a role in neuronal pathology. These findings implicate a loss of beta-III spectrin function in SCA5 pathogenesis and indicate that there are at least two physiological effects of beta-III spectrin loss that underpin a progressive loss of inhibitory cerebellar output, namely an intrinsic Purkinje cell membrane defect due to reduced sodium currents and alterations in glutamate signaling.


Subject(s)
Motor Activity/genetics , Purkinje Cells/metabolism , Purkinje Cells/pathology , Spectrin/deficiency , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Action Potentials/genetics , Animals , Atrophy/genetics , Cerebellum/pathology , Gait/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Spectrin/genetics , Spinocerebellar Ataxias/metabolism , Spinocerebellar Ataxias/physiopathology , Tremor/genetics
9.
Br J Haematol ; 149(5): 785-94, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20346007

ABSTRACT

Hereditary Spherocytosis (HS) is a haemolytic anaemia caused by erythrocyte protein membrane defects - spectrin, ankyrin, band 3 or protein 4.2 - that lead to membrane destabilization. This study aimed to evaluate the prevalence of protein deficiencies and the role of membrane proteins or membrane-linked proteins in membrane disturbance and in HS clinical outcome. A total of 215 Portuguese individuals were studied - 203 from 71 families plus 12 individual unrelated subjects; 160 of them were diagnosed with HS. They were classified as presenting mild, moderate or severe forms of HS according to the degree of haemolytic anaemia. Standardized electrophoretic erythrocyte membrane protein analysis was used to identify and quantify protein deficiencies. Band 3 and ankyrin were found to account for the majority of the erythrocyte protein defects underlying HS. Increasing isolated protein deficiency or increasing imbalance between combined protein deficiencies seemed to underlie HS severity, by increasing membrane destabilization. There was an increased membrane linkage of the cytosolic proteins, glyceraldehyde-3-phosphate dehydrogenase and peroxiredoxin 2, and of denatured haemoglobin, suggesting that this linkage could interfere with membrane structure. Our data suggest that the quantification and the analysis of RBC membrane proteins may be helpful in predicting the clinical outcome of HS.


Subject(s)
Erythrocyte Membrane/metabolism , Membrane Proteins/deficiency , Spherocytosis, Hereditary/blood , Adult , Anion Exchange Protein 1, Erythrocyte/deficiency , Ankyrins/deficiency , Child , Electrophoresis, Polyacrylamide Gel , Hemoglobins/metabolism , Homeodomain Proteins/blood , Humans , Prognosis , Severity of Illness Index , Spectrin/deficiency
10.
Br J Haematol ; 148(3): 456-65, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20092464

ABSTRACT

Lu/BCAM, the unique erythroid receptor for laminin 511/521, interacts with the erythrocyte membrane skeleton through spectrin binding. It has been reported that Hereditary Spherocytosis red blood cells (HS RBC) exhibit increased adhesion to laminin. We investigated the role of Lu/BCAM-spectrin interaction in the RBC adhesion properties of 2 splenectomised HS patients characterized by 40% spectrin deficiency. Under physiological flow conditions, HS RBC exhibited an exaggerated adhesion to laminin that was completely abolished by soluble Lu/BCAM. Triton extraction experiments revealed that a greater fraction of Lu/BCAM was unlinked to the membrane skeleton of HS RBC, as compared to normal RBC. Disruption of the spectrin interaction site in Lu/BCAM expressed in the transfected K562 cell line resulted in a weakened interaction to the skeleton and an enhanced interaction to laminin. These results demonstrated that the adhesion of HS RBC to laminin was mediated by Lu/BCAM and that its interaction with the spectrin-based skeleton negatively regulated cell adhesion to laminin. Finally, the results of this study strongly suggest that the reinforced adhesiveness of spectrin-deficient HS RBC to laminin is partly brought about by an impaired interaction between Lu/BCAM and the membrane skeleton.


Subject(s)
Cell Adhesion Molecules/blood , Erythrocyte Membrane/metabolism , Laminin/blood , Lutheran Blood-Group System/blood , Spectrin/metabolism , Spherocytosis, Hereditary/blood , Cell Adhesion/physiology , Erythrocytes/metabolism , Female , Hemorheology , Humans , Infant, Newborn , K562 Cells , Male , Phosphorylation , Recombinant Proteins/metabolism , Spectrin/deficiency
11.
Front Immunol ; 11: 620046, 2020.
Article in English | MEDLINE | ID: mdl-33519826

ABSTRACT

STAT3 gain-of-function (GOF) mutations can be responsible for an incomplete phenotype mainly characterized by hematological autoimmunity, even in the absence of other organ autoimmunity, growth impairment, or severe infections. We hereby report a case with an incomplete form of STAT3 GOF intensified by a concomitant hereditary hematological disease, which misleads the diagnosis. The patient presented with lymphadenopathy, splenomegaly, hypogammaglobulinemia, and severe autoimmune hemolytic anemia (AIHA) with critical complications, including stroke. A Primary Immune Regulatory Disorders (PIRD) was suspected, and molecular analysis revealed a de novo STAT3 gain-of-function mutation. The response to multiple immune suppressive treatments was ineffective, and further investigations revealed a spectrin deficiency. Ultimately, hematopoietic stem cell transplantation from a matched unrelated donor was able to cure the patient. Our case shows an atypical presentation of STAT3 GOF associated with hereditary spherocytosis, and how achievement of a good long-term outcome depends on a strict clinical and laboratory monitoring, as well as on prompt therapeutic intervention.


Subject(s)
Agammaglobulinemia/genetics , Anemia, Hemolytic, Autoimmune/genetics , Gain of Function Mutation , Lymphoproliferative Disorders/genetics , STAT3 Transcription Factor/genetics , Spectrin/deficiency , Agammaglobulinemia/immunology , Age of Onset , Anemia, Hemolytic, Autoimmune/immunology , Child , Cyclosporine/adverse effects , Cyclosporine/pharmacokinetics , Cyclosporine/therapeutic use , Cytochrome P-450 CYP3A/genetics , Female , Germ-Line Mutation , Graft vs Host Disease/drug therapy , Graft vs Host Disease/etiology , Growth Disorders/genetics , Hematopoietic Stem Cell Transplantation , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunosuppressive Agents/therapeutic use , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/immunology , Mutation, Missense , Polymorphism, Single Nucleotide , Prednisolone/therapeutic use , Retinal Hemorrhage/chemically induced , STAT3 Transcription Factor/physiology , Spectrin/genetics , Unrelated Donors
12.
Elife ; 92020 05 19.
Article in English | MEDLINE | ID: mdl-32425157

ABSTRACT

Previously, we showed that a hierarchy of spectrin cytoskeletal proteins maintains nodal Na+ channels (Liu et al., 2020). Here, using mice lacking Ɵ1, Ɵ4, or Ɵ1/Ɵ4 spectrins, we show this hierarchy does not function at axon initial segments (AIS). Although Ɵ1 spectrin, together with AnkyrinR (AnkR), compensates for loss of nodal Ɵ4 spectrin, it cannot compensate at AIS. We show AnkR lacks the domain necessary for AIS localization. Whereas loss of Ɵ4 spectrin causes motor impairment and disrupts AIS, loss of Ɵ1 spectrin has no discernable effect on central nervous system structure or function. However, mice lacking both neuronal Ɵ1 and Ɵ4 spectrin show exacerbated nervous system dysfunction compared to mice lacking Ɵ1 or Ɵ4 spectrin alone, including profound disruption of AIS Na+ channel clustering, progressive loss of nodal Na+ channels, and seizures. These results further define the important role of AIS and nodal spectrins for nervous system function.


Subject(s)
Axon Initial Segment/metabolism , Carrier Proteins/metabolism , Hippocampus/metabolism , Microfilament Proteins/metabolism , Spectrin/metabolism , Voltage-Gated Sodium Channels/metabolism , Animals , Ankyrins/metabolism , Behavior, Animal , Carrier Proteins/genetics , Cells, Cultured , Female , Hippocampus/physiopathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Motor Activity , Protein Domains , Rotarod Performance Test , Seizures/genetics , Seizures/metabolism , Seizures/physiopathology , Spectrin/deficiency , Spectrin/genetics
13.
J Cell Biol ; 125(5): 1057-65, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8195289

ABSTRACT

A spectrin-based membrane skeleton is important for the stability and organization of the erythrocyte. To study the role of spectrin in cells that possess complex cytoskeletons, we have generated alpha-spectrin-deficient erythroleukemia cell lines from sph/sph mice. These cells contain beta-spectrin, but lack alpha-spectrin as determined by immunoblot and Northern blot analyses. The effects of alpha-spectrin deficiency are apparent in the cells' irregular shape and fragility in culture. Capping of membrane glycoproteins by fluorescent lectin or antibodies occurs more rapidly in sph/sph than in wild-type erythroleukemia cells, and the caps appear more concentrated. The data support the idea that spectrin plays an important role in organizing membrane structure and limiting the lateral mobility of integral membrane glycoproteins in cells other than mature erythrocytes.


Subject(s)
Immunologic Capping , Spectrin/deficiency , Spectrin/physiology , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Friend murine leukemia virus , H-2 Antigens/metabolism , Leukemia, Erythroblastic, Acute , Mice , Mice, Mutant Strains , Time Factors
14.
J Cell Biol ; 157(4): 665-77, 2002 May 13.
Article in English | MEDLINE | ID: mdl-11994313

ABSTRACT

A common feature of multicellular animals is the ubiquitous presence of the spectrin cytoskeleton. Although discovered over 30 yr ago, the function of spectrin in non-erythrocytes has remained elusive. We have found that the spc-1 gene encodes the only alpha spectrin gene in the Caenorhabditis elegans genome. During embryogenesis, alpha spectrin localizes to the cell membrane in most if not all cells, starting at the first cell stage. Interestingly, this localization is dependent on beta spectrin but not beta(Heavy) spectrin. Furthermore, analysis of spc-1 mutants indicates that beta spectrin requires alpha spectrin to be stably recruited to the cell membrane. Animals lacking functional alpha spectrin fail to complete embryonic elongation and die just after hatching. These mutant animals have defects in the organization of the hypodermal apical actin cytoskeleton that is required for elongation. In addition, we find that the process of elongation is required for the proper differentiation of the body wall muscle. Specifically, when compared with myofilaments in wild-type animals the myofilaments of the body wall muscle in mutant animals are abnormally oriented relative to the longitudinal axis of the embryo, and the body wall muscle cells do not undergo normal cell shape changes.


Subject(s)
Body Patterning/physiology , Caenorhabditis elegans/embryology , Cell Membrane/metabolism , Cytoskeleton/metabolism , Embryo, Nonmammalian/embryology , Muscles/embryology , Spectrin/deficiency , Amino Acid Sequence/genetics , Animal Structures/cytology , Animal Structures/embryology , Animal Structures/metabolism , Animals , Base Sequence/genetics , Basement Membrane/cytology , Basement Membrane/embryology , Basement Membrane/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Cell Membrane/ultrastructure , Cell Size/genetics , Cytoskeleton/ultrastructure , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/physiology , Genes, Lethal/genetics , Molecular Sequence Data , Muscles/cytology , Muscles/metabolism , Mutation/genetics , Phenotype , Spectrin/genetics
15.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31550236

ABSTRACT

Increased fibrosis is a characteristic remodeling response to biomechanical and neurohumoral stress and a determinant of cardiac mechanical and electrical dysfunction in disease. Stress-induced activation of cardiac fibroblasts (CFs) is a critical step in the fibrotic response, although the precise sequence of events underlying activation of these critical cells in vivo remain unclear. Here, we tested the hypothesis that a ƟIV-spectrin/STAT3 complex is essential for maintenance of a quiescent phenotype (basal nonactivated state) in CFs. We reported increased fibrosis, decreased cardiac function, and electrical impulse conduction defects in genetic and acquired mouse models of ƟIV-spectrin deficiency. Loss of ƟIV-spectrin function promoted STAT3 nuclear accumulation and transcriptional activity, and it altered gene expression and CF activation. Furthermore, we demonstrate that a quiescent phenotype may be restored in ƟIV-spectrin-deficient fibroblasts by expressing a ƟIV-spectrin fragment including the STAT3-binding domain or through pharmacological STAT3 inhibition. We found that in vivo STAT3 inhibition abrogates fibrosis and cardiac dysfunction in the setting of global ƟIV-spectrin deficiency. Finally, we demonstrate that fibroblast-specific deletion of ƟIV-spectrin is sufficient to induce fibrosis and decreased cardiac function. We propose that the ƟIV-spectrin/STAT3 complex is a determinant of fibroblast phenotype and fibrosis, with implications for remodeling response in cardiovascular disease (CVD).


Subject(s)
Cardiovascular Diseases/physiopathology , Fibroblasts/pathology , Heart Ventricles/pathology , STAT3 Transcription Factor/metabolism , Spectrin/deficiency , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Disease Models, Animal , Female , Fibrosis , Heart Ventricles/cytology , Heart Ventricles/physiopathology , Humans , Male , Mice , Mice, Knockout , STAT3 Transcription Factor/antagonists & inhibitors , Spectrin/genetics , Ventricular Remodeling
16.
Oncogene ; 26(50): 7103-10, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17546056

ABSTRACT

Transforming growth factor-beta (TGF-beta) signaling members, TGF-beta receptor type II (TBRII), Smad2, Smad4 and Smad adaptor, embryonic liver fodrin (ELF), are prominent tumor suppressors in gastrointestinal cancers. Here, we show that 40% of elf(+/-) mice spontaneously develop hepatocellular cancer (HCC) with markedly increased cyclin D1, cyclin-dependent kinase 4 (Cdk4), c-Myc and MDM2 expression. Reduced ELF but not TBRII, or Smad4 was observed in 8 of 9 human HCCs (P<0.017). ELF and TBRII are also markedly decreased in human HCC cell lines SNU-398 and SNU-475. Restoration of ELF and TBRII in SNU-398 cells markedly decreases cyclin D1 as well as hyperphosphorylated-retinoblastoma (hyperphosphorylated-pRb). Thus, we show that TGF-beta signaling and Smad adaptor ELF suppress human hepatocarcinogenesis, potentially through cyclin D1 deregulation. Loss of ELF could serve as a primary event in progression toward a fully transformed phenotype and could hold promise for new therapeutic approaches in human HCCs.


Subject(s)
Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/physiology , Cyclins/metabolism , Liver Neoplasms, Experimental/etiology , Microfilament Proteins/physiology , Signal Transduction/physiology , Spectrin/physiology , Transforming Growth Factor beta2/antagonists & inhibitors , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cyclin D , Cyclins/antagonists & inhibitors , Humans , Liver Neoplasms, Experimental/metabolism , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Phosphorylation , Receptors, Transforming Growth Factor beta/metabolism , Retinoblastoma/metabolism , Signal Transduction/genetics , Spectrin/deficiency , Spectrin/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
17.
Integr Biol (Camb) ; 10(8): 450-463, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30027970

ABSTRACT

Sensory neurons embedded in skin are responsible for the sense of touch. In humans and other mammals, touch sensation depends on thousands of diverse somatosensory neurons. By contrast, Caenorhabditis elegans nematodes have six gentle touch receptor neurons linked to simple behaviors. The classical touch assay uses an eyebrow hair to stimulate freely moving C. elegans, evoking evasive behavioral responses. This assay has led to the discovery of genes required for touch sensation, but does not provide control over stimulus strength or position. Here, we present an integrated system for performing automated, quantitative touch assays that circumvents these limitations and incorporates automated measurements of behavioral responses. The Highly Automated Worm Kicker (HAWK) unites a microfabricated silicon force sensor holding a glass bead forming the contact surface and video analysis with real-time force and position control. Using this system, we stimulated animals along the anterior-posterior axis and compared responses in wild-type and spc-1(dn) transgenic animals, which have a touch defect due to expression of a dominant-negative α-spectrin protein fragment. As expected from prior studies, delivering large stimuli anterior and posterior to the mid-point of the body evoked a reversal and a speed-up, respectively. The probability of evoking a response of either kind depended on stimulus strength and location; once initiated, the magnitude and quality of both reversal and speed-up behavioral responses were uncorrelated with stimulus location, strength, or the absence or presence of the spc-1(dn) transgene. Wild-type animals failed to respond when the stimulus was applied near the mid-point. These results show that stimulus strength and location govern the activation of a characteristic motor program and that the C. elegans body surface consists of two receptive fields separated by a gap.


Subject(s)
Caenorhabditis elegans/physiology , Animals , Animals, Genetically Modified , Behavior, Animal/physiology , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Computer Systems , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Physical Stimulation/instrumentation , Sensory Receptor Cells/physiology , Spectrin/deficiency , Spectrin/genetics , Spectrin/physiology , Touch/physiology
18.
Blood Rev ; 21(1): 1-20, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16730867

ABSTRACT

The red cell membrane is one of the best known membranes in terms of structure, function and genetic disorders. As any plasma membrane it mediates transport functions. It also provides the erythrocytes with their resilience and deformability. Many of the proteins and the genes performing these functions are known in great detail, although some disease-responsible genes are yet to be elucidated. Basic knowledge has shed light on important groups of genetic disorders. The latter include (i) the disorders of the red cell mechanics: hereditary spherocytosis, hereditary elliptocytosis and poikilocytosis, and (ii) the disorders of the passive flux of the monovalent cations across the membrane: the stomacytoses and allied conditions. Reciprocally, many information have come from genetics abnormalities. We will review the mutation-disease relationship. A number of points will be underscored: widespread weak alleles modulate the expression of the SPTA1 gene, encoding the alpha-chain of spectrin; mutations in the anion exchanger can give rise to an array of distinct nosological entities, including a renal condition; splenectomy is banned in the stomatocytoses; a variety of stomatocyosis is part of a pleiotropic syndrome that may includes perinatal fetal liquid effusions. The diagnosis, follow-up and treatment of the involved diseases have gradually improved.


Subject(s)
Anemia, Hemolytic, Congenital/genetics , Erythrocytes/pathology , Membrane Proteins/genetics , Anemia, Hemolytic, Congenital/diagnosis , Anemia, Hemolytic, Congenital/physiopathology , Ankyrins/deficiency , Ankyrins/genetics , Apoptosis , Carrier Proteins/genetics , Cell Membrane/physiology , Erythrocytes/cytology , Humans , Membrane Proteins/deficiency , Microfilament Proteins/genetics , Mutation , Spectrin/deficiency , Spectrin/genetics
19.
J Clin Invest ; 82(2): 617-23, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3403720

ABSTRACT

Whereas marked variations in the clinical manifestations of hereditary spherocytosis have long been recognized, we have only recently begun to define the molecular basis for this heterogeneity. An important unanswered question is whether decreased spectrin results in reduced membrane mechanical stability, and if this reduction in membrane mechanical stability can be related to in vivo surface area loss. Using the ektacytometer, we quantitated membrane surface area and stability in erythrocytes from 18 individuals with hereditary spherocytosis and deficiencies of spectrin (30-80% of normal spectrin level). Membrane mechanical stability was reduced and the magnitude of the reductions correlated with the spectrin content. Moreover, the reductions in mechanical stability correlated with in vivo loss of membrane surface area. These data indicate that decreased spectrin content results in reduced membrane mechanical stability and surface area loss in vivo. We conclude that partial deficiencies of spectrin, reductions in membrane mechanical stability, and loss of membrane surface area are directly related and are major features determining the heterogeneous clinical manifestations of hereditary spherocytosis.


Subject(s)
Erythrocyte Deformability , Erythrocyte Membrane/physiology , Spectrin/deficiency , Spherocytosis, Hereditary/blood , Biomechanical Phenomena , Erythrocyte Membrane/pathology , Humans , Osmolar Concentration , Spherocytosis, Hereditary/pathology , Surface Properties
20.
J Clin Invest ; 81(1): 133-41, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3335631

ABSTRACT

Hereditary spherocytosis is a common hemolytic anemia associated with deficiencies in spectrin, the principal structural protein of the erythrocyte membrane-skeleton. We have examined 20 different individuals from 10 spherocytosis kindreds and 2 elliptocytosis kindreds to determine the effects of different levels of spectrin deficiency on the viscoelastic properties of the erythrocyte membrane. Micropipettes were used to perform single-cell micromechanical measurements of approximately 1,000 individual cells to determine the membrane elastic shear modulus, the apparent membrane bending stiffness, and whole cell recovery time constant for the different cell populations. The membrane viscosity was calculated by the product of the shear modulus and the recovery time constant. Results show correlation between the fractional reduction in shear modulus and the fractional reduction in spectrin content (determined by spectrin radioimmunoassay) and spectrin density (determined by the ratios of spectrin to band 3 on electrophoresis gels) suggesting that membrane shear elasticity is directly proportional to the surface density of spectrin on the membrane (P less than 0.001). The apparent membrane bending stiffness is also reduced in proportion to the density of spectrin (P less than 0.001). The membrane viscosity is reduced relative to control (P less than 0.001), but the nature of the relationship between spectrin density and membrane viscosity is less clearly defined. These studies document striking relationships between partial deficiencies of erythrocyte spectrin and specific viscoelastic properties of the mutant membranes.


Subject(s)
Erythrocyte Deformability , Erythrocyte Membrane/physiology , Spectrin/deficiency , Spherocytosis, Hereditary/blood , Elasticity , Erythrocyte Membrane/metabolism , Female , Humans , Male , Rheology , Spectrin/physiology , Spherocytes/pathology , Spherocytosis, Hereditary/genetics , Spherocytosis, Hereditary/pathology , Time Factors , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL