1
|
Davison CA, Garcia D, Feng C, Hao H, Jorgensen EM, Hammarlund M. The neuron-intrinsic membrane skeleton is required for motor neuron integrity throughout lifespan. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639536. [PMID: 40060495 PMCID: PMC11888272 DOI: 10.1101/2025.02.23.639536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Axons experience physical stress throughout an organism's lifetime, and disruptions in axonal integrity are hallmarks of both neurodegenerative diseases and traumatic injuries. The spectrin-based membrane periodic skeleton (MPS) is proposed to have a crucial role in maintaining axonal strength, flexibility, and resilience. To investigate the importance of the intrinsic MPS for GABAergic motor neuron integrity in C. elegans, we employed the auxin-inducible degron system to degrade β-spectrin/UNC-70 in a cell-type specific and time-dependent manner. Degradation of β-spectrin from all neurons beginning at larval development resulted in widespread axon breakage and regeneration in VD/DD GABAergic motor neurons in both larval and adult animals. Similarly, targeted degradation of β-spectrin in GABA neurons alone resulted in extensive breakage. Moreover, we found that depleting β-spectrin from the mature nervous system also induced axon breaks. By contrast, epidermal β-spectrin was not required for axon integrity of VD/DD neurons. These findings demonstrate the cell-intrinsic importance of neuronal β-spectrin/UNC-70 for axon integrity both during development and in adulthood.
Collapse
Affiliation(s)
- Carrie Ann Davison
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Daniela Garcia
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Chengye Feng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Hongyan Hao
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Erik M Jorgensen
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, Salt Lake City, UT, USA
| | - Marc Hammarlund
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Lead Contact
| |
Collapse
|
2
|
Lanning NJ, Mancour L, Argetsinger LS, Archer S, Carter-Su C. Identification of βIIΣ1-Spectrin as a Binding Partner of the GH-regulated Human Obesity Scaffold Protein SH2B1. Endocrinology 2025; 166:bqaf003. [PMID: 39801013 PMCID: PMC12053369 DOI: 10.1210/endocr/bqaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Indexed: 02/07/2025]
Abstract
SH2B1β is a multifunctional scaffold protein that modulates cytoskeletal processes such as cellular motility and neurite outgrowth. To identify novel SH2B1β-interacting proteins involved in these processes, a yeast 2-hybrid assay was performed. The C-terminal 159 residues of the cytoskeleton structural protein, βIIΣ1-spectrin, interacted with the N-terminal 260 residues of SH2B1β, a region implicated in SH2B1β enhancement of cell motility and localization at the plasma membrane. The interaction between SH2B1β and βIIΣ1-spectrin (2205-2363) requires residues 1 through 150 in SH2B1β, with residues 105 through 120 playing a key role. While βIIΣ1-spectrin (2205-2363) was expressed throughout the cell, it colocalized with SH2B1β when coexpressed with SH2B1β mutants with varied intracellular localizations. The SH2B1β-βIIΣ1-spectrin (2205-2363) interaction impaired the ability of SH2B1β to enter the nucleus. A slightly larger βIIΣ1-spectrin fragment (2170-2363) with an intact pleckstrin homology domain localized primarily to the plasma membrane and cytoplasm, similar to SH2B1β. Similarly, full-length βIIΣ1-spectrin colocalized at the plasma membrane and cytoplasm with SH2B1β as well as the SH2B1β-regulated tyrosine kinase, JAK2. Phosphorylation of spectrins has been shown to regulate their localization and function. Coexpression of βIIΣ1-spectrin, JAK2, and SH2B1β resulted in SH2B1β-dependent tyrosyl phosphorylation of βIIΣ1-spectrin. Finally, stimulation with GH induced formation of an endogenous complex containing βII-spectrin, SH2B1, and JAK2 in 3T3-F442A cells and increased tyrosyl phosphorylation of βII-spectrin. Our results identify a novel interaction between SH2B1β, βIIΣ1-spectrin, and JAK2 resulting in JAK2- and SHB1-dependent tyrosyl phosphorylation of βII-spectrin. This raises the possibility that the many other ligand-activated tyrosine kinases that signal through SH2B1 form similar complexes with βIIΣ1-spectrin.
Collapse
Affiliation(s)
- Nathan J Lanning
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Liliya Mancour
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lawrence S Argetsinger
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Stephen Archer
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christin Carter-Su
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
3
|
Readnour BM, Tjia-Fleck S, McCann NR, Ayinuola YA, Castellino FJ. High-resolution cryo-EM analysis of a Streptococcus pyogenes M-protein/human plasminogen complex. Structure 2024; 32:2231-2243.e4. [PMID: 39500317 DOI: 10.1016/j.str.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/18/2024] [Accepted: 10/02/2024] [Indexed: 12/08/2024]
Abstract
The importance of human plasminogen (hPg)/plasmin (hPm)/cell receptor complexes in invasiveness of cells has been amply established. The objective of this investigation was to determine a high-resolution structure of a major Group A Streptococcus (GAS) bacterial receptor (PAM) for hPg/hPm when bound on a cell surface to its major ligand, hPg. As a model cell surface with endogenous PAM, we employed engineered PAM-expressing lentivirus (LV) particles. We show that the ectodomain of a PAM-type M-Protein (M-Prt), in complex with hPg, is folded through distinct intra- and inter-domain interactions to a more compact form on the cell surface, thus establishing a new paradigm for membrane-bound M-Prt/ligand structures. These studies provide a framework for addressing the need for treatments of GAS disease by providing a molecular platform to solve structures of virulence-determining membrane proteins.
Collapse
Affiliation(s)
- Bradley M Readnour
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sheiny Tjia-Fleck
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Nathan R McCann
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yetunde A Ayinuola
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Francis J Castellino
- W.M. Keck Center for Transgene Research, University of Notre Dame, Notre Dame, IN 46556, USA; Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
4
|
Denha SA, DeLaet NR, Abukamil AW, Alexopoulos AN, Keller AR, Atang AE, Avery AW. Molecular consequences of SCA5 mutations in the spectrin-repeat domains of β-III-spectrin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613313. [PMID: 39345584 PMCID: PMC11429872 DOI: 10.1101/2024.09.17.613313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Spinocerebellar ataxia type 5 (SCA5) mutations in the protein β-III-spectrin cluster to the N-terminal actin-binding domain (ABD) and the central spectrin-repeat domains (SRDs). We previously reported that a common molecular consequence of ABD-localized SCA5 mutations is increased actin binding. However, little is known about the molecular consequences of the SRD-localized mutations. It is known that the SRDs of β-spectrin proteins interact with α-spectrin to form an α/β-spectrin dimer. In addition, it is known that SRDs neighbouring the β-spectrin ABD enhance actin binding. Here, we tested the impact of the SRD-localized R480W and the E532_M544del mutations on the binding of β-III-spectrin to α-II-spectrin and actin. Using multiple experimental approaches, we show that both the R480W and E532_M544del mutants can bind α-II-spectrin. However, E532_M544del causes partial uncoupling of complementary SRDs in the α/β-spectrin dimer. Further, the R480W mutant forms large intracellular inclusions when co-expressed with α-II-spectrin in cells, supporting that R480W mutation grossly disrupts the α-II/β-III-spectrin physical complex. Moreover, actin-binding assays show that E532_M544del, but not R480W, increases β-III-spectrin actin binding. Altogether, these data support that SRD-localized mutations alter key interactions of β-III-spectrin with α-II-spectrin and actin.
Collapse
Affiliation(s)
- Sarah A. Denha
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| | - Naomi R. DeLaet
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| | - Abeer W. Abukamil
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| | | | - Amanda R. Keller
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| | - Alexandra E. Atang
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| | - Adam W. Avery
- Department of Chemistry, Oakland University, Rochester, MI 48309-4479, USA
| |
Collapse
|
5
|
Boehler JF, Brown KJ, Beatka M, Gonzalez JP, Donisa Dreghici R, Soustek-Kramer M, McGonigle S, Ganot A, Palmer T, Lowie C, Chamberlain JS, Lawlor MW, Morris CA. Clinical potential of microdystrophin as a surrogate endpoint. Neuromuscul Disord 2023; 33:40-49. [PMID: 36575103 DOI: 10.1016/j.nmd.2022.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Accelerated approval based on a likely surrogate endpoint can be life-changing for patients suffering from a rare progressive disease with unmet medical need, as it substantially hastens access to potentially lifesaving therapies. In one such example, antisense morpholinos were approved to treat Duchenne muscular dystrophy (DMD) based on measurement of shortened dystrophin in skeletal muscle biopsies as a surrogate biomarker. New, promising therapeutics for DMD include AAV gene therapy to restore another form of dystrophin termed mini- or microdystrophin. AAV-microdystrophins are currently in clinical trials but have yet to be accepted by regulatory agencies as reasonably likely surrogate endpoints. To evaluate microdystrophin expression as a reasonably likely surrogate endpoint for DMD, this review highlights dystrophin biology in the context of functional and clinical benefit to support the argument that microdystrophin proteins have a high probability of providing clinical benefit based on their rational design. Unlike exon-skipping based strategies, the approach of rational design allows for functional capabilities (i.e. quality) of the protein to be maximized with every patient receiving the same optimized microdystrophin. Therefore, the presence of rationally designed microdystrophin in a muscle biopsy is likely to predict clinical benefit and is consequently a strong candidate for a surrogate endpoint analysis to support accelerated approval.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Kristy J Brown
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Margaret Beatka
- Diverge TSL, 247 Freshwater Way Suite 610, Milwaukee, WI 53204, United States
| | - J Patrick Gonzalez
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | | | | | - Sharon McGonigle
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Annie Ganot
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Timothy Palmer
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Caitlin Lowie
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, United States
| | - Michael W Lawlor
- Diverge TSL, 247 Freshwater Way Suite 610, Milwaukee, WI 53204, United States
| | - Carl A Morris
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States.
| |
Collapse
|
6
|
Hale J, An X, Guo X, Gao E, Papoin J, Blanc L, Hillyer CD, Gratzer W, Baines A, Mohandas N. αI-spectrin represents evolutionary optimization of spectrin for red blood cell deformability. Biophys J 2021; 120:3588-3599. [PMID: 34352252 PMCID: PMC8456306 DOI: 10.1016/j.bpj.2021.07.027] [Citation(s) in RCA: 4] [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: 03/22/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/15/2022] Open
Abstract
Spectrin tetramers of the membranes of enucleated mammalian erythrocytes play a critical role in red blood cell survival in circulation. One of the spectrins, αI, emerged in mammals with enucleated red cells after duplication of the ancestral α-spectrin gene common to all animals. The neofunctionalized αI-spectrin has moderate affinity for βI-spectrin, whereas αII-spectrin, expressed in nonerythroid cells, retains ancestral characteristics and has a 10-fold higher affinity for βI-spectrin. It has been hypothesized that this adaptation allows for rapid make and break of tetramers to accommodate membrane deformation. We have tested this hypothesis by generating mice with high-affinity spectrin tetramers formed by exchanging the site of tetramer formation in αI-spectrin (segments R0 and R1) for that of αII-spectrin. Erythrocytes with αIIβI presented normal hematologic parameters yet showed increased thermostability, and their membranes were significantly less deformable; under low shear forces, they displayed tumbling behavior rather than tank treading. The membrane skeleton is more stable with αIIβI and shows significantly less remodeling under deformation than red cell membranes of wild-type mice. These data demonstrate that spectrin tetramers undergo remodeling in intact erythrocytes and that this is required for the normal deformability of the erythrocyte membrane. We conclude that αI-spectrin represents evolutionary optimization of tetramer formation: neither higher-affinity tetramers (as shown here) nor lower affinity (as seen in hemolytic disease) can support the membrane properties required for effective tissue oxygenation in circulation.
Collapse
Affiliation(s)
- John Hale
- The Red Cell Physiology Laboratory, The New York Blood Center, New York, New York.
| | - Xiuli An
- Membrane Biology Laboratory, The New York Blood Center, New York, New York
| | - Xinhua Guo
- Membrane Biology Laboratory, The New York Blood Center, New York, New York
| | - Erjing Gao
- The Red Cell Physiology Laboratory, The New York Blood Center, New York, New York
| | - Julien Papoin
- Nelkin Laboratory of Pediatric Oncology and Laboratory of Developmental Erythropoiesis, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Lionel Blanc
- Nelkin Laboratory of Pediatric Oncology and Laboratory of Developmental Erythropoiesis, The Feinstein Institutes for Medical Research, Manhasset, New York; Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | | | - Walter Gratzer
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Anthony Baines
- Department of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Narla Mohandas
- The Red Cell Physiology Laboratory, The New York Blood Center, New York, New York
| |
Collapse
|
7
|
Tutwiler V. To deform or not to deform: the evolutionary basis of mammalian red blood cell deformability. Biophys J 2021; 120:3539-3540. [PMID: 34403646 DOI: 10.1016/j.bpj.2021.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 10/20/2022] Open
Affiliation(s)
- Valerie Tutwiler
- Department of Biomedical Engineering, Rutgers University, New Brunswick, New Jersey.
| |
Collapse
|
8
|
Cusseddu R, Robert A, Côté JF. Strength Through Unity: The Power of the Mega-Scaffold MACF1. Front Cell Dev Biol 2021; 9:641727. [PMID: 33816492 PMCID: PMC8012552 DOI: 10.3389/fcell.2021.641727] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/23/2021] [Indexed: 12/26/2022] Open
Abstract
The tight coordination of diverse cytoskeleton elements is required to support several dynamic cellular processes involved in development and tissue homeostasis. The spectraplakin-family of proteins are composed of multiple domains that provide versatility to connect different components of the cytoskeleton, including the actin microfilaments, microtubules and intermediates filaments. Spectraplakins act as orchestrators of precise cytoskeletal dynamic events. In this review, we focus on the prototypical spectraplakin MACF1, a protein scaffold of more than 700 kDa that coordinates the crosstalk between actin microfilaments and microtubules to support cell-cell connections, cell polarity, vesicular transport, proliferation, and cell migration. We will review over two decades of research aimed at understanding the molecular, physiological and pathological roles of MACF1, with a focus on its roles in developmental and cancer. A deeper understanding of MACF1 is currently limited by technical challenges associated to the study of such a large protein and we discuss ideas to advance the field.
Collapse
Affiliation(s)
- Rebecca Cusseddu
- Montreal Clinical Research Institute, Montreal, QC, Canada
- Molecular Biology Programs, Université de Montréal, Montreal, QC, Canada
| | - Amélie Robert
- Montreal Clinical Research Institute, Montreal, QC, Canada
| | - Jean-François Côté
- Montreal Clinical Research Institute, Montreal, QC, Canada
- Molecular Biology Programs, Université de Montréal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Negretti NM, Gourley CR, Talukdar PK, Clair G, Klappenbach CM, Lauritsen CJ, Adkins JN, Konkel ME. The Campylobacter jejuni CiaD effector co-opts the host cell protein IQGAP1 to promote cell entry. Nat Commun 2021; 12:1339. [PMID: 33637714 PMCID: PMC7910587 DOI: 10.1038/s41467-021-21579-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Campylobacter jejuni is a foodborne pathogen that binds to and invades the epithelial cells lining the human intestinal tract. Maximal invasion of host cells by C. jejuni requires cell binding as well as delivery of the Cia proteins (Campylobacter invasion antigens) to the host cell cytosol via the flagellum. Here, we show that CiaD binds to the host cell protein IQGAP1 (a Ras GTPase-activating-like protein), thus displacing RacGAP1 from the IQGAP1 complex. This, in turn, leads to the unconstrained activity of the small GTPase Rac1, which is known to have roles in actin reorganization and internalization of C. jejuni. Our results represent the identification of a host cell protein targeted by a flagellar secreted effector protein and demonstrate that C. jejuni-stimulated Rac signaling is dependent on IQGAP1.
Collapse
Affiliation(s)
- Nicholas M Negretti
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Christopher R Gourley
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Prabhat K Talukdar
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Geremy Clair
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Courtney M Klappenbach
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Cody J Lauritsen
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Joshua N Adkins
- Integrative Omics, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Michael E Konkel
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
| |
Collapse
|
10
|
Abstract
Cooperativity is a hallmark of protein folding, but the thermodynamic origins of cooperativity are difficult to quantify. Tandem repeat proteins provide a unique experimental system to quantify cooperativity due to their internal symmetry and their tolerance of deletion, extension, and in some cases fragmentation into single repeats. Analysis of repeat proteins of different lengths with nearest-neighbor Ising models provides values for repeat folding ([Formula: see text]) and inter-repeat coupling (ΔGi-1,i). In this article, we review the architecture of repeat proteins and classify them in terms of ΔGi and ΔGi-1,i; this classification scheme groups repeat proteins according to their degree of cooperativity. We then present various statistical thermodynamic models, based on the 1D-Ising model, for analysis of different classes of repeat proteins. We use these models to analyze data for highly and moderately cooperative and noncooperative repeat proteins and relate their fitted parameters to overall structural features.
Collapse
Affiliation(s)
- Mark Petersen
- Program in Molecular Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, USA.,T.C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| | - Doug Barrick
- T.C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| |
Collapse
|
11
|
Barbarino F, Wäschenbach L, Cavalho-Lemos V, Dillenberger M, Becker K, Gohlke H, Cortese-Krott MM. Targeting spectrin redox switches to regulate the mechanoproperties of red blood cells. Biol Chem 2020; 402:317-331. [PMID: 33544503 DOI: 10.1515/hsz-2020-0293] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022]
Abstract
The mechanical properties of red blood cells (RBCs) are fundamental for their physiological role as gas transporters. RBC flexibility and elasticity allow them to survive the hemodynamic changes in the different regions of the vascular tree, to dynamically contribute to the flow thereby decreasing vascular resistance, and to deform during the passage through narrower vessels. RBC mechanoproperties are conferred mainly by the structural characteristics of their cytoskeleton, which consists predominantly of a spectrin scaffold connected to the membrane via nodes of actin, ankyrin and adducin. Changes in redox state and treatment with thiol-targeting molecules decrease the deformability of RBCs and affect the structure and stability of the spectrin cytoskeleton, indicating that the spectrin cytoskeleton may contain redox switches. In this perspective review, we revise current knowledge about the structural and functional characterization of spectrin cysteine redox switches and discuss the current lines of research aiming to understand the role of redox regulation on RBC mechanical properties. These studies may provide novel functional targets to modulate RBC function, blood viscosity and flow, and tissue perfusion in disease conditions.
Collapse
Affiliation(s)
- Frederik Barbarino
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Lucas Wäschenbach
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Virginia Cavalho-Lemos
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| | - Melissa Dillenberger
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, D-35392, Giessen, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, D-35392, Giessen, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, D-52425, Jülich, Germany
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich Heine University, Postfach 128, Universitätsstrasse 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
12
|
Bose D, Chakrabarti A. Multiple Functions of Spectrin: Convergent Effects. J Membr Biol 2020; 253:499-508. [PMID: 32990795 DOI: 10.1007/s00232-020-00142-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/19/2020] [Indexed: 10/23/2022]
Abstract
Spectrin is a multifunctional, multi-domain protein most well known in the membrane skeleton of mature human erythrocytes. Here we review the literature on the crosstalk of the chaperone activity of spectrin with its other functionalities. We hypothesize that the chaperone activity is derived from the surface exposed hydrophobic patches present in individual "spectrin-repeat" domains and show a competition between the membrane phospholipid binding functionality and chaperone activity of spectrin. Moreover, we show that post-translational modifications such as glycation which shield these surface exposed hydrophobic patches, reduce the chaperone function. On the other hand, oligomerization which is linked to increase of hydrophobicity is seen to increase it. We note that spectrin seems to prefer haemoglobin as its chaperone client, binding with it preferentially over other denatured proteins. Spectrin is also known to interact with unstable haemoglobin variants with a higher affinity than in the case of normal haemoglobin. We propose that chaperone activity of spectrin could be important in the cellular biochemistry of haemoglobin, particularly in the context of diseases.
Collapse
Affiliation(s)
- Dipayan Bose
- Crystallography & Molecular Biology Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India.,Homi Bhabha National Institute, Mumbai, 400094, India
| | - Abhijit Chakrabarti
- Crystallography & Molecular Biology Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata, 700064, India. .,Homi Bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
13
|
Nagao S, Suda A, Kobayashi H, Shibata N, Higuchi Y, Hirota S. Thermodynamic Control of Domain Swapping by Modulating the Helical Propensity in the Hinge Region of Myoglobin. Chem Asian J 2020; 15:1743-1749. [DOI: 10.1002/asia.202000307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/20/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Satoshi Nagao
- Division of Materials ScienceGraduate School of Science and TechnologyNara Institute of Science and Technology 8916-5 Takayama Ikoma Nara 630-0192 Japan
- Present address: Graduate School of Life ScienceUniversity of Hyogo 3-2-1 Koto Kamigori-cho, Ako-gun Hyogo 678-1297 Japan
| | - Ayaka Suda
- Division of Materials ScienceGraduate School of Science and TechnologyNara Institute of Science and Technology 8916-5 Takayama Ikoma Nara 630-0192 Japan
| | - Hisashi Kobayashi
- Division of Materials ScienceGraduate School of Science and TechnologyNara Institute of Science and Technology 8916-5 Takayama Ikoma Nara 630-0192 Japan
| | - Naoki Shibata
- Graduate School of Life ScienceUniversity of Hyogo 3-2-1 Koto Kamigori-cho, Ako-gun Hyogo 678-1297 Japan
| | - Yoshiki Higuchi
- Graduate School of Life ScienceUniversity of Hyogo 3-2-1 Koto Kamigori-cho, Ako-gun Hyogo 678-1297 Japan
| | - Shun Hirota
- Division of Materials ScienceGraduate School of Science and TechnologyNara Institute of Science and Technology 8916-5 Takayama Ikoma Nara 630-0192 Japan
| |
Collapse
|
14
|
Asaro RJ, Zhu Q. Vital erythrocyte phenomena: what can theory, modeling, and simulation offer? Biomech Model Mechanobiol 2020; 19:1361-1388. [DOI: 10.1007/s10237-020-01302-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
|
15
|
Mechanical Unfolding of Spectrin Repeats Induces Water-Molecule Ordering. Biophys J 2020; 118:1076-1089. [PMID: 32027822 DOI: 10.1016/j.bpj.2020.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/24/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanical processes are involved at many stages of the development of living cells, and often external forces applied to a biomolecule result in its unfolding. Although our knowledge of the unfolding mechanisms and the magnitude of the forces involved has evolved, the role that water molecules play in the mechanical unfolding of biomolecules has not yet been fully elucidated. To this end, we investigated with steered molecular dynamics simulations the mechanical unfolding of dystrophin's spectrin repeat 1 and related the changes in the protein's structure to the ordering of the surrounding water molecules. Our results indicate that upon mechanically induced unfolding of the protein, the solvent molecules become more ordered and increase their average number of hydrogen bonds. In addition, the unfolded structures originating from mechanical pulling expose an increasing amount of the hydrophobic residues to the solvent molecules, and the uncoiled regions adapt a convex surface with a small radius of curvature. As a result, the solvent molecules reorganize around the protein's small protrusions in structurally ordered waters that are characteristic of the so-called "small-molecule regime," which allows water to maintain a high hydrogen bond count at the expense of an increased structural order. We also determined that the response of water to structural changes in the protein is localized to the specific regions of the protein that undergo unfolding. These results indicate that water plays an important role in the mechanically induced unfolding of biomolecules. Our findings may prove relevant to the ever-growing interest in understanding macromolecular crowding in living cells and their effects on protein folding, and suggest that the hydration layer may be exploited as a means for short-range allosteric communication.
Collapse
|
16
|
Cheng C, Hu Z, Cao L, Peng C, He Y. The scavenger receptor SCARA1 (CD204) recognizes dead cells through spectrin. J Biol Chem 2019; 294:18881-18897. [PMID: 31653705 DOI: 10.1074/jbc.ra119.010110] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/22/2019] [Indexed: 01/05/2023] Open
Abstract
Scavenger receptor class A member 1 (SCARA1 or CD204) is an immune receptor highly expressed on macrophages. It forms homotrimers on the cell surface and plays important roles in regulating immune responses via its involvement in multiple pathways. However, both the structure and the functional roles of SCARA1 are not fully understood. Here, we determined the crystal structure of the C-terminal SRCR domain of SCARA1 at 1.8 Å resolution, revealing its Ca2+-binding site. Results from cell-based assays revealed that SCARA1 can recognize dead cells, rather than live cells, specifically through its SRCR domain and in a Ca2+-dependent manner. Furthermore, by combining MS and biochemical assays, we found that cellular spectrin is the binding target of SCARA1 on dead cells and that the SRCR domain of SCARA1 recognizes the SPEC repeats of spectrin in the presence of Ca2+ We also found that macrophages can internalize dead cells or debris from both erythrocytes and other cells through the interaction between SCARA1 and spectrin, suggesting that SCARA1 could function as a scavenging receptor that recognizes dead cells. These results suggest that spectrin, which is one of the major components of the cytoskeleton, acts as a cellular marker that enables the recognition of dead cells by the immune system.
Collapse
Affiliation(s)
- Chen Cheng
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenzheng Hu
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Longxing Cao
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chao Peng
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yongning He
- State Key Laboratory of Molecular Biology, National Center for Protein Science Shanghai, Shanghai Science Research Center, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
17
|
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
|
18
|
Machek ML, Sonnenschein HA, Graham SI, Shikwana F, Kim SL, Garcia DuBar S, Minzer ID, Wey R, Bell JK. Predicting and validating a model of suppressor of IKKepsilon through biophysical characterization. Protein Sci 2019; 28:1423-1436. [PMID: 31074891 PMCID: PMC6635840 DOI: 10.1002/pro.3640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/03/2019] [Accepted: 05/01/2019] [Indexed: 12/23/2022]
Abstract
Suppressor of IKKepsilon (SIKE) is a 207 residue protein that is implicated in the TLR3-TANK-binding kinase-1-mediated response to viral infection. SIKE's function in this pathway is unknown, but SIKE forms interactions with two distinct cytoskeletal proteins, α-actinin and tubulin, and SIKE knockout reduces cell migration. As structure informs function and in the absence of solved structural homologs, our studies were directed toward creating a structural model of SIKE through biochemical and biophysical characterization to probe and interrogate SIKE function. Circular dichroism revealed a primarily (73%) helical structure of minimal stability ( =32°C) but reversibly denatured. Limited proteolysis (LP) and chemical modification identified the N-terminal 2/3 of the protein as dynamic and accessible, whereas size exclusion chromatography (SEC) confirmed three homo-oligomeric species. SEC coupled to chemical crosslinking characterized the primary species as dimeric, a secondary hexameric species, and a higher order aggregate/polymer. Fluorescence polarization using intrinsic tryptophan fluorescence contextualized the anisotropy value for the SIKE dimer (molecular weight 51.8 kDa) among proteins of known structure, bovine serum albumin (BSA; 66 kDa), and glutamate dehydrogenase (GDH; 332 kDa). Radii of gyration for BSA and GDH provided exclusionary values for SIKE tertiary and dimeric quaternary models that otherwise conformed to secondary structure, LP, and modification data. Dimeric quaternary models were further culled using acrylamide quenching data of SIKE's single tryptophan that showed a single, protected environment. The low cooperativity of folding and regions of dynamic and potentially disordered structure advance the hypothesis that SIKE forms a conformational ensemble of native states that accommodate SIKE's interactions with multiple, distinct protein-binding partners.
Collapse
Affiliation(s)
- Megan L. Machek
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| | - Halie A. Sonnenschein
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| | - Sasha‐Kaye I. Graham
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| | - Flowreen Shikwana
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| | | | | | - Ian D. Minzer
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| | - Ryan Wey
- ACS SEED ScholarsUniversity of San DiegoSan DiegoCalifornia92110
| | - Jessica K. Bell
- Department of Chemistry & BiochemistryUniversity of San DiegoSan DiegoCalifornia92110
| |
Collapse
|
19
|
Mias-Lucquin D, Chéron A, Le Rumeur E, Hubert JF, Delalande O. Fine mapping of hydrophobic contacts reassesses the organization of the first three dystrophin coiled-coil repeats. Protein Sci 2018; 28:561-570. [PMID: 30468271 DOI: 10.1002/pro.3557] [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: 07/09/2018] [Revised: 10/29/2018] [Accepted: 11/18/2018] [Indexed: 11/12/2022]
Abstract
Coiled-coil domain is a structural motif found in proteins crucial for achievement of central biological processes, such as cellular cohesion or neuro-transmission. The coiled-coil fold consists of alpha-helices bundle that can be repeated to form larger filament. Hydrophobic residues, distributed following a regular seven-residues' pattern, named heptad pattern, are commonly admitted to be essential for the formation and the stability of canonical coiled-coil repeats. Here we investigated the first three coiled-coil repeats (R1-R3) of the central domain of dystrophin, a scaffolding protein in muscle cells whose deficiency leads to Duchenne and Becker Muscular Dystrophies. By an atomic description of the hydrophobic interactions, we highlighted (i) that coiled-coil filament conformational changes are associated to specific patterns of inter-helices hydrophobic contacts, (ii) that inter-repeat hydrophobic interactions determine the behavior of linker regions including filament kinks, and (iii) that a non-strict conservation of the heptad patterns is leading to a relative plasticity of the dystrophin coiled-coil repeats. These structural features and modulations of the coiled-coil fold could better explain the mechanical properties of the central domain of dystrophin. This contribution to the understanding of the structure-function relationship of dystrophin, and especially of the R1-R3 fragment frequently used in the design of protein for gene therapies, should help in the improvement of the strategies for the cure of muscular dystrophies.
Collapse
|
20
|
Muthu Krishnan S. Using Chou's general PseAAC to analyze the evolutionary relationship of receptor associated proteins (RAP) with various folding patterns of protein domains. J Theor Biol 2018; 445:62-74. [DOI: 10.1016/j.jtbi.2018.02.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/24/2018] [Accepted: 02/12/2018] [Indexed: 01/31/2023]
|
21
|
Takahashi H, Rico F, Chipot C, Scheuring S. α-Helix Unwinding as Force Buffer in Spectrins. ACS NANO 2018; 12:2719-2727. [PMID: 29390177 DOI: 10.1021/acsnano.7b08973] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Spectrins are cytoskeletal proteins located at the inner face of the plasma membrane, making connections between membrane anchors and the actin cortex, and between actin filaments. Spectrins share a common structure forming a bundle of 3 α-helices and play a major role during cell deformation. Here, we used high-speed force spectroscopy and steered molecular dynamics simulations to understand the mechanical stability of spectrin, revealing a molecular force buffering function. We find that spectrin acts as a soft spring at short extensions (70-100 Å). Under continuous external stretching, its α-helices unwind, leading to a viscous mechanical response over larger extensions (100-300 Å), represented by a constant-force plateau in force/extension curves. This viscous force buffering emerges from a quasi-equilibrium competition between disruption and re-formation of α-helical hydrogen bonds. Our results suggest that, in contrast to β-sheet proteins, which unfold in a catastrophic event, α-helical spectrins dominantly unwind, providing a viscous force buffer over extensions about 5 times their folded length.
Collapse
Affiliation(s)
- Hirohide Takahashi
- U1006 INSERM, Université Aix-Marseille, Parc Scientifique et Technologique de Luminy , 163 Avenue de Luminy , 13009 Marseille , France
- Department of Anesthesiology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
- Departments of Physiology and Biophysics , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| | - Felix Rico
- U1006 INSERM, Université Aix-Marseille, Parc Scientifique et Technologique de Luminy , 163 Avenue de Luminy , 13009 Marseille , France
| | - Christophe Chipot
- Laboratoire International Associé Centre National de la Recherche Scientifique et University of Illinois at Urbana-Champaign , UMR 7565, Université de Lorraine, B.P. 70239, 54506 Vandœuvre-lès-Nancy Cedex, France, and Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, Urbana , Illinois 61801 , United States
| | - Simon Scheuring
- U1006 INSERM, Université Aix-Marseille, Parc Scientifique et Technologique de Luminy , 163 Avenue de Luminy , 13009 Marseille , France
- Department of Anesthesiology , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
- Departments of Physiology and Biophysics , Weill Cornell Medicine , 1300 York Avenue , New York , New York 10065 , United States
| |
Collapse
|
22
|
Hu L, Huang Z, Wu Z, Ali A, Qian A. Mammalian Plakins, Giant Cytolinkers: Versatile Biological Functions and Roles in Cancer. Int J Mol Sci 2018; 19:ijms19040974. [PMID: 29587367 PMCID: PMC5979291 DOI: 10.3390/ijms19040974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
Cancer is a highly lethal disease that is characterized by aberrant cell proliferation, migration, and adhesion, which are closely related to the dynamic changes of cytoskeletons and cytoskeletal-adhesion. These will further result in cell invasion and metastasis. Plakins are a family of giant cytolinkers that connect cytoskeletal elements with each other and to junctional complexes. With various isoforms composed of different domain structures, mammalian plakins are broadly expressed in numerous tissues. They play critical roles in many cellular processes, including cell proliferation, migration, adhesion, and signaling transduction. As these cellular processes are key steps in cancer development, mammalian plakins have in recent years attracted more and more attention for their potential roles in cancer. Current evidence shows the importance of mammalian plakins in various human cancers and demonstrates mammalian plakins as potential biomarkers for cancer. Here, we introduce the basic characteristics of mammalian plakins, review the recent advances in understanding their biological functions, and highlight their roles in human cancers, based on studies performed by us and others. This will provide researchers with a comprehensive understanding of mammalian plakins, new insights into the development of cancer, and novel targets for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Lifang Hu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zizhan Huang
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Zixiang Wu
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Arshad Ali
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Airong Qian
- Laboratory for Bone Metabolism, Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
23
|
White JT, Toptygin D, Cohen R, Murphy N, Hilser VJ. Structural Stability of the Coiled-Coil Domain of Tumor Susceptibility Gene (TSG)-101. Biochemistry 2017; 56:4646-4655. [PMID: 28776372 PMCID: PMC5616090 DOI: 10.1021/acs.biochem.7b00469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
![]()
The
tumor susceptibility gene-101 coiled coil domain (TSG101cc)
is an integral component of the endosomal maturation machinery and
cytokinesis, and also interacts with several transcription factors.
The TSG101cc has been crystallized as a homotetramer but is known
to interact with two of its binding partners as a heterotrimer. To
investigate this apparent discrepancy, we examined the solution thermodynamics
of the TSG101cc. Here, we use circular dichroism, differential scanning
calorimetry, analytical ultracentrifugation, fluorescence, and structural
thermodynamic analysis to investigate the structural stability and
the unfolding of the TSG101cc. We demonstrate that TSG101cc exists
in solution primarily as a tetramer, which unfolds in a two-state
manner. Surprisingly, no homodimeric or homotrimeric species were
detected. Structural thermodynamic analysis of the homotetrameric
structure and comparison with known oligomeric coiled-coils suggests
that the TSG101cc homotetramer is comparatively unstable on a per
residue basis. Furthermore, the homotrimeric coiled-coil is predicted
to be much less stable than the functional heterotrimeric coiled-coil
in the endosomal sorting complex required for transport 1 (ESCRT1).
These results support a model whereby the tetramer–monomer
equilibrium of TSG101 serves as the cellular reservoir of TSG101,
which is effectively outcompeted when its binding partners are present
and the heteroternary complex can form.
Collapse
Affiliation(s)
- Jordan T White
- Department of Biology, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Dmitri Toptygin
- Department of Biology, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Randy Cohen
- Department of Biology, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Natalie Murphy
- Department of Biology, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Vincent J Hilser
- Department of Biology, Johns Hopkins University , 3400 North Charles Street, Baltimore, Maryland 21218, United States
| |
Collapse
|
24
|
Bonjack-Shterengartz M, Avnir D. The enigma of the near-symmetry of proteins: Domain swapping. PLoS One 2017; 12:e0180030. [PMID: 28708874 PMCID: PMC5510828 DOI: 10.1371/journal.pone.0180030] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/08/2017] [Indexed: 01/25/2023] Open
Abstract
The majority of proteins form oligomers which have rotational symmetry. Literature has suggested many functional advantages that the symmetric packing offers. Yet, despite these advantages, the vast majority of protein oligomers are only nearly symmetric. A key question in the field of proteins structure is therefore, if symmetry is so advantageous, why do oligomers settle for aggregates that do not maximize that structural property? The answer to that question is apparently multi-parametric, and involves distortions at the interaction zones of the monomer units of the oligomer in order to minimize the free energy, the dynamics of the protein, the effects of surroundings parameters, and the mechanism of oligomerization. The study of this problem is in its infancy: Only the first parameter has been explored so far. Here we focus on the last parameter-the mechanism of formation. To test this effect we have selected to focus on the domain swapping mechanism of oligomerization, by which oligomers form in a mechanism that swaps identical portions of monomeric units, resulting in an interwoven oligomer. We are using continuous symmetry measures to analyze in detail the oligomer formed by this mechanism, and found, that without exception, in all analyzed cases, perfect symmetry is given away, and we are able to identify that the main burden of distortion lies in the hinge regions that connect the swapped portions. We show that the continuous symmetry analysis method clearly identifies the hinge region of swapped domain proteins-considered to be a non-trivial task. We corroborate our conclusion about the central role of the hinge region in affecting the symmetry of the oligomers, by a special probability analysis developed particularly for that purpose.
Collapse
Affiliation(s)
- Maayan Bonjack-Shterengartz
- Institute of Chemistry and the Lise Meitner Minerva Center for Computational Quantum Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Avnir
- Institute of Chemistry and the Lise Meitner Minerva Center for Computational Quantum Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
25
|
Gessner C, Steinchen W, Bédard S, J Skinner J, Woods VL, Walsh TJ, Bange G, Pantazatos DP. Computational method allowing Hydrogen-Deuterium Exchange Mass Spectrometry at single amide Resolution. Sci Rep 2017. [PMID: 28630467 PMCID: PMC5476592 DOI: 10.1038/s41598-017-03922-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Hydrogen-deuterium exchange (HDX) coupled with mass spectrometry (HDXMS) is a rapid and effective method for localizing and determining protein stability and dynamics. Localization is routinely limited to a peptide resolution of 5 to 20 amino acid residues. HDXMS data can contain information beyond that needed for defining protein stability at single amide resolution. Here we present a method for extracting this information from an HDX dataset to generate a HDXMS protein stability fingerprint. High resolution (HR)-HDXMS was applied to the analysis of a model protein of a spectrin tandem repeat that exemplified an intuitive stability profile based on the linkage of two triple helical repeats connected by a helical linker. The fingerprint recapitulated expected stability maximums and minimums with interesting structural features that corroborate proposed mechanisms of spectrin flexibility and elasticity. HR-HDXMS provides the unprecedented ability to accurately assess protein stability at the resolution of a single amino acid. The determination of HDX stability fingerprints may be broadly applicable in many applications for understanding protein structure and function as well as protein ligand interactions.
Collapse
Affiliation(s)
- Chris Gessner
- Indiana University, Department of Informatics and Computing, Bloomington, IN, USA
| | - Wieland Steinchen
- Philipps-University Marburg, Faculty of Chemistry & LOEWE Center for Synthetic Microbiology Hans-Meerwein-Strasse, 35043, Marburg, Germany
| | - Sabrina Bédard
- GlaxoSmithKline, Platform Technology & Science, Collegeville Road, Collegeville, Pennsylvania, 19426, United States
| | - John J Skinner
- iHuman Institute, ShanghaiTech University, 99 Haike Road, Pudong, Shanghai, China
| | - Virgil L Woods
- Indiana University, Department of Informatics and Computing, Bloomington, IN, USA
| | - Thomas J Walsh
- Weill Cornell Medicine, Transplantation-Oncology Infectious Disease Program, Division of Infectious Diseases, 1300 York Ave, New York, NY, 10065, USA
| | - Gert Bange
- Philipps-University Marburg, Faculty of Chemistry & LOEWE Center for Synthetic Microbiology Hans-Meerwein-Strasse, 35043, Marburg, Germany
| | - Dionysios P Pantazatos
- Weill Cornell Medicine, Transplantation-Oncology Infectious Disease Program, Division of Infectious Diseases, 1300 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Moffat JJ, Ka M, Jung EM, Smith AL, Kim WY. The role of MACF1 in nervous system development and maintenance. Semin Cell Dev Biol 2017; 69:9-17. [PMID: 28579452 DOI: 10.1016/j.semcdb.2017.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/12/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Microtubule-actin crosslinking factor 1 (MACF1), also known as actin crosslinking factor 7 (ACF7), is essential for proper modulation of actin and microtubule cytoskeletal networks. Most MACF1 isoforms are expressed broadly in the body, but some are exclusively found in the nervous system. Consequentially, MACF1 is integrally involved in multiple neural processes during development and in adulthood, including neurite outgrowth and neuronal migration. Furthermore, MACF1 participates in several signaling pathways, including the Wnt/β-catenin and GSK-3 signaling pathways, which regulate key cellular processes, such as proliferation and cell migration. Genetic mutation or dysregulation of the MACF1 gene has been associated with neurodevelopmental and neurodegenerative diseases, specifically schizophrenia and Parkinson's disease. MACF1 may also play a part in neuromuscular disorders and have a neuroprotective role in the optic nerve. In this review, the authors seek to synthesize recent findings relating to the roles of MACF1 within the nervous system and explore potential novel functions of MACF1 not yet examined.
Collapse
Affiliation(s)
- Jeffrey J Moffat
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Minhan Ka
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Eui-Man Jung
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Amanda L Smith
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Woo-Yang Kim
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
27
|
Bose D, Patra M, Chakrabarti A. Effect of pH on stability, conformation, and chaperone activity of erythroid & non-erythroid spectrin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2017; 1865:694-702. [PMID: 28373029 DOI: 10.1016/j.bbapap.2017.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 11/29/2022]
Abstract
Spectrin, a major component of the eukaryotic membrane skeleton, has been shown to have chaperone like activity. Here we investigate the pH induced changes in the structure and stability of erythroid and brain spectrin by spectroscopic methods. We also correlate these changes with modulations of chaperone potential at different pH. We have followed the pH induced structural changes by circular dichroism spectroscopy and intrinsic tryptophan fluorescence. It is seen that lowering the pH from 9 has little effect on structure of the proteins till about pH6. At pH4, there is significant change of the secondary structure of the proteins, along with a 5nm hypsochromic shift of the emission maxima. Below pH4 the proteins undergo acid denaturation. Probing exposed hydrophobic patches on the proteins using protein-bound 8-anilinonaphthalene-1-sulfonate fluorescence demonstrates that there is higher solvent accessibility of hydrophobic surfaces in both forms of spectrin at around pH4. Dynamic light scattering and 90° light scattering studies show that the both forms of spectrin forms oligomers at pH~4. Chemical unfolding data shows that these oligomers are less stable than the tetrameric form. Aggregation studies with BSA show that at pH4, both spectrins exhibit better chaperone activity. This enhancement of chaperone like activity appears to result from an increase in regions of solvent-exposed hydrophobicity and oligomeric state of the spectrins which in turn are induced by moderately acid pH. This may have in-vivo implications in cells facing stress conditions where cytoplasmic pH is lowered.
Collapse
Affiliation(s)
- Dipayan Bose
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, HBNI, Kolkata, India
| | - Malay Patra
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, HBNI, Kolkata, India
| | - Abhijit Chakrabarti
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, HBNI, Kolkata, India.
| |
Collapse
|
28
|
Delalande O, Czogalla A, Hubert JF, Sikorski A, Le Rumeur E. Dystrophin and Spectrin, Two Highly Dissimilar Sisters of the Same Family. Subcell Biochem 2017; 82:373-403. [PMID: 28101868 DOI: 10.1007/978-3-319-49674-0_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dystrophin and Spectrin are two proteins essential for the organization of the cytoskeleton and for the stabilization of membrane cells. The comparison of these two sister proteins, and with the dystrophin homologue utrophin, enables us to emphasise that, despite a similar topology with common subdomains and a common structural basis of a three-helix coiled-coil, they show a large range of dissimilarities in terms of genetics, cell expression and higher level structural organisation. Interactions with cellular partners, including proteins and membrane phospholipids, also show both strikingly similar and very different behaviours. The differences between dystrophin and spectrin are also illustrated by the large variety of pathological anomalies emerging from the dysfunction or the absence of these proteins, showing that they are keystones in their function of providing a scaffold that sustains cell structure.
Collapse
Affiliation(s)
- Olivier Delalande
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France.
| | - Aleksander Czogalla
- Biotechnology Faculty, Department of Cytobiochemistry, University of Wrocław, ul. joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jean-François Hubert
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France
| | - Aleksander Sikorski
- Biotechnology Faculty, Department of Cytobiochemistry, University of Wrocław, ul. joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Elisabeth Le Rumeur
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France
| |
Collapse
|
29
|
Hu L, Su P, Li R, Yin C, Zhang Y, Shang P, Yang T, Qian A. Isoforms, structures, and functions of versatile spectraplakin MACF1. BMB Rep 2016; 49:37-44. [PMID: 26521939 PMCID: PMC4914211 DOI: 10.5483/bmbrep.2016.49.1.185] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Indexed: 11/20/2022] Open
Abstract
Spectraplakins are crucially important communicators, linking cytoskeletal components to each other and cellular junctions. Microtubule actin crosslinking factor 1 (MACF1), also known as actin crosslinking family 7 (ACF7), is a member of the spectraplakin family. It is expressed in numerous tissues and cells as one extensively studied spectraplakin. MACF1 has several isoforms with unique structures and well-known function to be able to crosslink F-actin and microtubules. MACF1 is one versatile spectraplakin with various functions in cell processes, embryo development, tissue-specific functions, and human diseases. The importance of MACF1 has become more apparent in recent years. Here, we summarize the current knowledge on the presence and function of MACF1 and provide perspectives on future research of MACF1 based on our studies and others. [BMB Reports 2016; 49(1): 37-44]
Collapse
Affiliation(s)
- Lifang Hu
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peihong Su
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Runzhi Li
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chong Yin
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Yan Zhang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Peng Shang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Tuanmin Yang
- Honghui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P. R. China
| | - Airong Qian
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| |
Collapse
|
30
|
Calkins S, Youssef NH. Insights into the Utility of the Focal Adhesion Scaffolding Proteins in the Anaerobic Fungus Orpinomyces sp. C1A. PLoS One 2016; 11:e0163553. [PMID: 27685796 PMCID: PMC5042518 DOI: 10.1371/journal.pone.0163553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/11/2016] [Indexed: 12/30/2022] Open
Abstract
Focal adhesions (FAs) are large eukaryotic multiprotein complexes that are present in all metazoan cells and function as stable sites of tight adhesion between the extracellular matrix (ECM) and the cell's cytoskeleton. FAs consist of anchor membrane protein (integrins), scaffolding proteins (e.g. α-actinin, talin, paxillin, and vinculin), signaling proteins of the IPP complex (e.g. integrin-linked kinase, α-parvin, and PINCH), and signaling kinases (e.g. focal adhesion kinase (FAK) and Src kinase). While genes encoding complete focal adhesion machineries are present in genomes of all multicellular Metazoa; incomplete machineries were identified in the genomes of multiple non-metazoan unicellular Holozoa, basal fungal lineages, and amoebozoan representatives. Since a complete FA machinery is required for functioning, the putative role, if any, of these incomplete FA machineries is currently unclear. We sought to examine the expression patterns of FA-associated genes in the anaerobic basal fungal isolate Orpinomyces sp. strain C1A under different growth conditions and at different developmental stages. Strain C1A lacks clear homologues of integrin, and the two signaling kinases FAK and Src, but encodes for all scaffolding proteins, and the IPP complex proteins. We developed a protocol for synchronizing growth of C1A cultures, allowing for the collection and mRNA extraction from flagellated spores, encysted germinating spores, active zoosporangia, and late inactive sporangia of strain C1A. We demonstrate that the genes encoding the FA scaffolding proteins α-actinin, talin, paxillin, and vinculin are indeed transcribed under all growth conditions, and at all developmental stages of growth. Further, analysis of the observed transcriptional patterns suggests the putative involvement of these components in alternative non-adhesion-specific functions, such as hyphal tip growth during germination and flagellar assembly during zoosporogenesis. Based on these results, we propose putative alternative functions for such proteins in the anaerobic gut fungi. Our results highlight the presumed diverse functionalities of FA scaffolding proteins in basal fungi.
Collapse
Affiliation(s)
- Shelby Calkins
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078, United States of America
| | - Noha H. Youssef
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, OK 74078, United States of America
| |
Collapse
|
31
|
HDInsight4PSi: Boosting performance of 3D protein structure similarity searching with HDInsight clusters in Microsoft Azure cloud. Inf Sci (N Y) 2016. [DOI: 10.1016/j.ins.2016.02.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Ivanov IT, Paarvanova B. Dielectric relaxations on erythrocyte membrane as revealed by spectrin denaturation. Bioelectrochemistry 2016; 110:59-68. [PMID: 27071054 DOI: 10.1016/j.bioelechem.2016.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/25/2016] [Accepted: 03/26/2016] [Indexed: 11/30/2022]
Abstract
We studied the effect of spectrin denaturation at 49.5°C (TA) on the dielectric relaxations and related changes in the complex impedance, Z*, complex capacitance, C*, and dielectric loss curve of suspensions containing human erythrocytes, erythrocyte ghost membranes (EMs) and Triton-X-100 residues of EMs. The loss curve prior to, minus the loss curve after TA, resulted in a bell-shaped peak at 1.5MHz. The changes in the real and imaginary components of Z* and C* at TA, i.e., ΔZre, ΔZim, ΔCre and ΔCim, calculated in the same way, strongly varied with frequency. Between 1.0 and 12MHz the -ΔZim vs ΔZre, and ΔCim vs ΔCre plots depicted semicircles with critical frequencies, fcr, at 2.5MHz expressing recently reported relaxation of spectrin dipoles. Between 0.02 and 1.0MHz the -ΔZim vs ΔZre plot exhibited another relaxation whose fcr mirrored that of beta relaxation. This relaxation was absent on Triton-X-shells, while on erythrocytes and EMs it was inhibited by selective dissociation of either attachment sites between spectrin and bilayer. Considering above findings and inaccessibility of cytosole to outside field at such frequencies, the latter relaxation was assumed originating from a piezoelectric effect on the highly deformable spectrin filaments.
Collapse
Affiliation(s)
- I T Ivanov
- Dept. of Physics, Biophysics, Reontgenology and Radiology, Medical Faculty, Thracian University, Stara Zagora 6000, Bulgaria.
| | - B Paarvanova
- Dept. of Physics, Biophysics, Reontgenology and Radiology, Medical Faculty, Thracian University, Stara Zagora 6000, Bulgaria
| |
Collapse
|
33
|
Triplett JC, Swomley AM, Kirk J, Grimes KM, Lewis KN, Orr ME, Rodriguez KA, Cai J, Klein JB, Buffenstein R, Butterfield DA. Reaching Out to Send a Message: Proteins Associated with Neurite Outgrowth and Neurotransmission are Altered with Age in the Long-Lived Naked Mole-Rat. Neurochem Res 2016; 41:1625-34. [DOI: 10.1007/s11064-016-1877-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 02/06/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
|
34
|
Ying J, Lin C, Wu J, Guo L, Qiu T, Ling Y, Shan L, Zhou H, Zhao D, Wang J, Liang J, Zhao J, Jiao Y, Lu N, Zhao H. Anaplastic Lymphoma Kinase Rearrangement in Digestive Tract Cancer: Implication for Targeted Therapy in Chinese Population. PLoS One 2015; 10:e0144731. [PMID: 26678488 PMCID: PMC4683076 DOI: 10.1371/journal.pone.0144731] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 11/23/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Anaplastic lymphoma kinase (ALK) rearrangements define a subgroup of lung cancer which is eligible to targeted kinase inhibition. The aim of this study is to observe the incidence rate of ALK fusion in a large cohort of Chinese digestive tract cancer patients. PATIENTS AND METHODS Tissue microarray (TMA) was constructed from 808 digestive tract cancer cases, including 169 esophageal squamous cell carcinoma, 182 gastric cancer and 457 colorectal cancer (CRC) cases. We tested all cases for ALK expression via a fully automated immunohistochemistry (IHC) assay. The IHC-positive cases were subjected to fluorescence in situ hybridization (FISH), real-time polymerase chain reaction (qRT-PCR), target gene enrichment and sequencing for confirmation of ALK gene rearrangement and discovery of novel fusion partner. RESULTS Among the tested cases, 2 (0.44%) CRC cases showed positive both by IHC and FISH. By qRT-PCR, EML4-ALK fusion was found in one IHC-positive CRC case. In another IHC-positive CRC case, target gene enrichment and sequencing revealed ALK was fused to a novel partner, spectrin beta non-erythrocytic 1 (SPTBN1). One gastric cancer case showed partially positive IHC result, but no fusion was found by FISH and gene sequencing. CONCLUSIONS The incidence rate of ALK gene fusion in Chinese CRC patients was 0.44%,but not detectable in gastric and esophageal cancers. The novel SPTBN1 -ALK fusion, together with other ALK fusion genes, may become a potential target for anti-ALK therapy.
Collapse
Affiliation(s)
- Jianming Ying
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Chen Lin
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Jian Wu
- MyGenostics Inc., Baltimore, MD, United States of America
| | - Lei Guo
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Tian Qiu
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Yun Ling
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Ling Shan
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Haitao Zhou
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Dongbing Zhao
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Jian Wang
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Jianwei Liang
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Jianjun Zhao
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Yuchen Jiao
- Laboratory of Cell and Molecular Biology & State Key Laboratory of Molecular Oncology, Cancer Institute & Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- * E-mail: (HZ); (NL); (YCJ)
| | - Ning Lu
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
- * E-mail: (HZ); (NL); (YCJ)
| | - Hong Zhao
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, PR China
- * E-mail: (HZ); (NL); (YCJ)
| |
Collapse
|
35
|
An Adaptable Spectrin/Ankyrin-Based Mechanism for Long-Range Organization of Plasma Membranes in Vertebrate Tissues. CURRENT TOPICS IN MEMBRANES 2015; 77:143-84. [PMID: 26781832 DOI: 10.1016/bs.ctm.2015.10.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ankyrins are membrane-associated proteins that together with their spectrin partners are responsible for micron-scale organization of vertebrate plasma membranes, including those of erythrocytes, excitable membranes of neurons and heart, lateral membrane domains of columnar epithelial cells, and striated muscle. Ankyrins coordinate functionally related membrane transporters and cell adhesion proteins (15 protein families identified so far) within plasma membrane compartments through independently evolved interactions of intrinsically disordered sequences with a highly conserved peptide-binding groove formed by the ANK repeat solenoid. Ankyrins are coupled to spectrins, which are elongated organelle-sized proteins that form mechanically resilient arrays through cross-linking by specialized actin filaments. In addition to protein interactions, cellular targeting and assembly of spectrin/ankyrin domains also critically depend on palmitoylation of ankyrin-G by aspartate-histidine-histidine-cysteine 5/8 palmitoyltransferases, as well as interaction of beta-2 spectrin with phosphoinositide lipids. These lipid-dependent spectrin/ankyrin domains are not static but are locally dynamic and determine membrane identity through opposing endocytosis of bulk lipids as well as specific proteins. A partnership between spectrin, ankyrin, and cell adhesion molecules first emerged in bilaterians over 500 million years ago. Ankyrin and spectrin may have been recruited to plasma membranes from more ancient roles in organelle transport. The basic bilaterian spectrin-ankyrin toolkit markedly expanded in vertebrates through gene duplications combined with variation in unstructured intramolecular regulatory sequences as well as independent evolution of ankyrin-binding activity by ion transporters involved in action potentials and calcium homeostasis. In addition, giant vertebrate ankyrins with specialized roles in axons acquired new coding sequences by exon shuffling. We speculate that early axon initial segments and epithelial lateral membranes initially were based on spectrin-ankyrin-cell adhesion molecule assemblies and subsequently served as "incubators," where ion transporters independently acquired ankyrin-binding activity through positive selection.
Collapse
|
36
|
Anatomy of the red cell membrane skeleton: unanswered questions. Blood 2015; 127:187-99. [PMID: 26537302 DOI: 10.1182/blood-2014-12-512772] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/30/2015] [Indexed: 11/20/2022] Open
Abstract
The red cell membrane skeleton is a pseudohexagonal meshwork of spectrin, actin, protein 4.1R, ankyrin, and actin-associated proteins that laminates the inner membrane surface and attaches to the overlying lipid bilayer via band 3-containing multiprotein complexes at the ankyrin- and actin-binding ends of spectrin. The membrane skeleton strengthens the lipid bilayer and endows the membrane with the durability and flexibility to survive in the circulation. In the 36 years since the first primitive model of the red cell skeleton was proposed, many additional proteins have been discovered, and their structures and interactions have been defined. However, almost nothing is known of the skeleton's physiology, and myriad questions about its structure remain, including questions concerning the structure of spectrin in situ, the way spectrin and other proteins bind to actin, how the membrane is assembled, the dynamics of the skeleton when the membrane is deformed or perturbed by parasites, the role lipids play, and variations in membrane structure in unique regions like lipid rafts. This knowledge is important because the red cell membrane skeleton is the model for spectrin-based membrane skeletons in all cells, and because defects in the red cell membrane skeleton underlie multiple hemolytic anemias.
Collapse
|
37
|
Jan A, Basit S, Wakil SM, Ramzan K, Ahmad W. A novel homozygous variant in the dsp gene underlies the first case of non-syndromic form of alopecia. Arch Dermatol Res 2015; 307:793-801. [DOI: 10.1007/s00403-015-1590-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/31/2015] [Accepted: 06/29/2015] [Indexed: 02/04/2023]
|
38
|
Brown JW, Bullitt E, Sriswasdi S, Harper S, Speicher DW, McKnight CJ. The Physiological Molecular Shape of Spectrin: A Compact Supercoil Resembling a Chinese Finger Trap. PLoS Comput Biol 2015; 11:e1004302. [PMID: 26067675 PMCID: PMC4466138 DOI: 10.1371/journal.pcbi.1004302] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/27/2015] [Indexed: 01/29/2023] Open
Abstract
The primary, secondary, and tertiary structures of spectrin are reasonably well defined, but the structural basis for the known dramatic molecular shape change, whereby the molecular length can increase three-fold, is not understood. In this study, we combine previously reported biochemical and high-resolution crystallographic data with structural mass spectroscopy and electron microscopic data to derive a detailed, experimentally-supported quaternary structure of the spectrin heterotetramer. In addition to explaining spectrin’s physiological resting length of ~55-65 nm, our model provides a mechanism by which spectrin is able to undergo a seamless three-fold extension while remaining a linear filament, an experimentally observed property. According to the proposed model, spectrin’s quaternary structure and mechanism of extension is similar to a Chinese Finger Trap: at shorter molecular lengths spectrin is a hollow cylinder that extends by increasing the pitch of each spectrin repeat, which decreases the internal diameter. We validated our model with electron microscopy, which demonstrated that, as predicted, spectrin is hollow at its biological resting length of ~55-65 nm. The model is further supported by zero-length chemical crosslink data indicative of an approximately 90 degree bend between adjacent spectrin repeats. The domain-domain interactions in our model are entirely consistent with those present in the prototypical linear antiparallel heterotetramer as well as recently reported inter-strand chemical crosslinks. The model is consistent with all known physical properties of spectrin, and upon full extension our Chinese Finger Trap Model reduces to the ~180-200 nm molecular model currently in common use. Spectrins are cytoskeletal and scaffolding proteins ubiquitously expressed in essentially all cell-types. Despite unequivocal evidence for a short physiological length of ~55–65 nm at rest, spectrin is typically represented as an extended ~200 nm molecule that is implied based on crystallographic structures of a number of tandem repeats. Here, we incorporate previously reported biochemical and crystallographic data with structural mass spectroscopy and electron microscopic data to derive a detailed, experimentally-supported quaternary structure of the physiological compact form of spectrin. In addition to explaining spectrin’s physiological resting length (~55–65 nm), our model provides a mechanism by which spectrin can undergo a seamless three-fold extension, which is an experimentally observed property that is responsible for restoration of cell shape after mechanical deformation.
Collapse
Affiliation(s)
- Jeffrey W. Brown
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Internal Medicine Residency Program, University of Pittsburgh Medical Center, UPMC Montefiore Hospital, Pittsburgh, Pennsylvania, United States of America
| | - Esther Bullitt
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Sira Sriswasdi
- Center for Systems and Computational Biology, and Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, Pennsylvania, United States of America
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sandra Harper
- Center for Systems and Computational Biology, and Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - David W. Speicher
- Center for Systems and Computational Biology, and Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, Pennsylvania, United States of America
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - C. James McKnight
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
39
|
Ngo KX, Kodera N, Katayama E, Ando T, Uyeda TQP. Cofilin-induced unidirectional cooperative conformational changes in actin filaments revealed by high-speed atomic force microscopy. eLife 2015; 4. [PMID: 25642645 PMCID: PMC4337605 DOI: 10.7554/elife.04806] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 01/02/2015] [Indexed: 01/12/2023] Open
Abstract
High-speed atomic force microscopy was employed to observe structural changes in actin filaments induced by cofilin binding. Consistent with previous electron and fluorescence microscopic studies, cofilin formed clusters along actin filaments, where the filaments were 2-nm thicker and the helical pitch was ∼25% shorter, compared to control filaments. Interestingly, the shortened helical pitch was propagated to the neighboring bare zone on the pointed-end side of the cluster, while the pitch on the barbed-end side was similar to the control. Thus, cofilin clusters induce distinctively asymmetric conformational changes in filaments. Consistent with the idea that cofilin favors actin structures with a shorter helical pitch, cofilin clusters grew unidirectionally toward the pointed-end of the filament. Severing was often observed near the boundaries between bare zones and clusters, but not necessarily at the boundaries. DOI:http://dx.doi.org/10.7554/eLife.04806.001 Actin is one of the most abundant proteins found inside all eukaryotic cells including plant, animal, and fungal cells. This protein is involved in a wide range of biological processes that are essential for an organism's survival. Actin proteins form long filaments that help cells to maintain their shape and also provide the force required for cells to divide and/or move. Actin filaments are helical in shape and are made up of many actin subunits joined together. Actin filaments are changeable structures that continuously grow and shrink as new actin subunits are added to or removed from the ends of the filaments. One end of an actin filament grows faster than the other; the fast-growing end is known as the barbed-end, while the slow-growing end is referred to as the pointed-end. Over 100 other proteins are known to bind to and work with actin to regulate its roles in cells and how it forms into filaments. Cofilin is one such protein that binds to and forms clusters on actin filaments and it can also sever actin filaments. Severing an actin filament can encourage the filament to disassemble, or it can help produce new barbed ends that can then grow into new filaments. Previous work had suggested that cofilin severs actin filaments at the junction between regions on the filament that are coated with cofilin and those that are not. It was also known that cofilin binding to a filament causes the filament to change shape, and that the shape change is propagated to neighboring sections of the filaments not coated with cofilin. However, the details of where cofilin binds and how changes in shape are propagated along an actin filament were not known. Furthermore, the findings of these previous studies were largely based on examining still images of actin filaments, which are unlike the constantly changing filaments of living cells. Ngo, Kodera et al. have now analyzed what happens when cofilin binds to and forms clusters along actin filaments using a recently developed imaging technique called high-speed atomic force microscopy. This technique can be used to directly visualize individual proteins in action. Consistent with previous findings, Ngo, Kodera et al. observed that filaments coated with cofilin are thicker than those filaments without cofilin; and that cofilin binding also substantially reduces the helical twist of the filament. Ngo, Kodera et al. also found that these changes in shape are propagated along the filament but in only one direction—towards the pointed-end. Moreover, cofilin clusters also only grew towards the pointed-end of the actin filament—and the filaments were often severed near, but not exactly at, the junctions between cofilin-coated and uncoated regions. Such one-directional changes in shape of the actin filaments presumably help to regulate how other actin binding proteins can interact with the filament and consequently regulate the roles of the filaments themselves. DOI:http://dx.doi.org/10.7554/eLife.04806.002
Collapse
Affiliation(s)
- Kien Xuan Ngo
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Noriyuki Kodera
- Department of Physics and Bio-AFM Frontier Research Center, Kanazawa University, Kanazawa, Japan
| | - Eisaku Katayama
- Department of Biology, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Toshio Ando
- Department of Physics and Bio-AFM Frontier Research Center, Kanazawa University, Kanazawa, Japan
| | - Taro Q P Uyeda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| |
Collapse
|
40
|
Patra M, Mukhopadhyay C, Chakrabarti A. Probing conformational stability and dynamics of erythroid and nonerythroid spectrin: effects of urea and guanidine hydrochloride. PLoS One 2015; 10:e0116991. [PMID: 25617632 PMCID: PMC4305312 DOI: 10.1371/journal.pone.0116991] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/18/2014] [Indexed: 11/19/2022] Open
Abstract
We have studied the conformational stability of the two homologous membrane skeletal proteins, the erythroid and non-erythroid spectrins, in their dimeric and tetrameric forms respectively during unfolding in the presence of urea and guanidine hydrochloride (GuHCl). Fluorescence and circular dichroism (CD) spectroscopy have been used to study the changes of intrinsic tryptophan fluorescence, anisotropy, far UV-CD and extrinsic fluorescence of bound 1-anilinonapthalene-8-sulfonic acid (ANS). Chemical unfolding of both proteins were reversible and could be described as a two state transition. The folded erythroid spectrin and non-erythroid spectrin were directly converted to unfolded monomer without formation of any intermediate. Fluorescence quenching, anisotropy, ANS binding and dynamic light scattering data suggest that in presence of low concentrations of the denaturants (up-to 1M) hydrogen bonding network and van der Waals interaction play a role inducing changes in quaternary as well as tertiary structures without complete dissociation of the subunits. This is the first report of two large worm like, multi-domain proteins obeying twofold rule which is commonly found in small globular proteins. The free energy of stabilization (ΔGuH20) for the dimeric spectrin has been 20 kcal/mol lesser than the tetrameric from.
Collapse
Affiliation(s)
- Malay Patra
- Chemistry Department, University of Calcutta, Kolkata, West Bengal, India
| | | | - Abhijit Chakrabarti
- Crystallography & Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, West Bengal, India
| |
Collapse
|
41
|
Reconstructing genome-wide protein-protein interaction networks using multiple strategies with homologous mapping. PLoS One 2015; 10:e0116347. [PMID: 25602759 PMCID: PMC4300222 DOI: 10.1371/journal.pone.0116347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/08/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND One of the crucial steps toward understanding the biological functions of a cellular system is to investigate protein-protein interaction (PPI) networks. As an increasing number of reliable PPIs become available, there is a growing need for discovering PPIs to reconstruct PPI networks of interesting organisms. Some interolog-based methods and homologous PPI families have been proposed for predicting PPIs from the known PPIs of source organisms. RESULTS Here, we propose a multiple-strategy scoring method to identify reliable PPIs for reconstructing the mouse PPI network from two well-known organisms: human and fly. We firstly identified the PPI candidates of target organisms based on homologous PPIs, sharing significant sequence similarities (joint E-value ≤ 1 × 10(-40)), from source organisms using generalized interolog mapping. These PPI candidates were evaluated by our multiple-strategy scoring method, combining sequence similarities, normalized ranks, and conservation scores across multiple organisms. According to 106,825 PPI candidates in yeast derived from human and fly, our scoring method can achieve high prediction accuracy and outperform generalized interolog mapping. Experiment results show that our multiple-strategy score can avoid the influence of the protein family size and length to significantly improve PPI prediction accuracy and reflect the biological functions. In addition, the top-ranked and conserved PPIs are often orthologous/essential interactions and share the functional similarity. Based on these reliable predicted PPIs, we reconstructed a comprehensive mouse PPI network, which is a scale-free network and can reflect the biological functions and high connectivity of 292 KEGG modules, including 216 pathways and 76 structural complexes. CONCLUSIONS Experimental results show that our scoring method can improve the predicting accuracy based on the normalized rank and evolutionary conservation from multiple organisms. Our predicted PPIs share similar biological processes and cellular components, and the reconstructed genome-wide PPI network can reflect network topology and modularity. We believe that our method is useful for inferring reliable PPIs and reconstructing a comprehensive PPI network of an interesting organism.
Collapse
|
42
|
Nicolas A, Delalande O, Hubert JF, Le Rumeur E. The spectrin family of proteins: A unique coiled-coil fold for various molecular surface properties. J Struct Biol 2014; 186:392-401. [DOI: 10.1016/j.jsb.2014.03.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/12/2014] [Accepted: 03/14/2014] [Indexed: 01/11/2023]
|
43
|
Kwa LG, Wensley BG, Alexander CG, Browning SJ, Lichman BR, Clarke J. The folding of a family of three-helix bundle proteins: spectrin R15 has a robust folding nucleus, unlike its homologous neighbours. J Mol Biol 2014; 426:1600-10. [PMID: 24373753 PMCID: PMC3988883 DOI: 10.1016/j.jmb.2013.12.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/13/2013] [Accepted: 12/17/2013] [Indexed: 11/13/2022]
Abstract
Three homologous spectrin domains have remarkably different folding characteristics. We have previously shown that the slow-folding R16 and R17 spectrin domains can be altered to resemble the fast folding R15, in terms of speed of folding (and unfolding), landscape roughness and folding mechanism, simply by substituting five residues in the core. Here we show that, by contrast, R15 cannot be engineered to resemble R16 and R17. It is possible to engineer a slow-folding version of R15, but our analysis shows that this protein neither has a rougher energy landscape nor does change its folding mechanism. Quite remarkably, R15 appears to be a rare example of a protein with a folding nucleus that does not change in position or in size when its folding nucleus is disrupted. Thus, while two members of this protein family are remarkably plastic, the third has apparently a restricted folding landscape.
Collapse
Affiliation(s)
- Lee Gyan Kwa
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Beth G Wensley
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Crispin G Alexander
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Stuart J Browning
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Benjamin R Lichman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Jane Clarke
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
44
|
Spectrin and phospholipids - the current picture of their fascinating interplay. Cell Mol Biol Lett 2014; 19:158-79. [PMID: 24569979 PMCID: PMC6276000 DOI: 10.2478/s11658-014-0185-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 02/19/2014] [Indexed: 12/02/2022] Open
Abstract
The spectrin-based membrane skeleton is crucial for the mechanical stability and resilience of erythrocytes. It mainly contributes to membrane integrity, protein organization and trafficking. Two transmembrane protein macro-complexes that are linked together by spectrin tetramers play a crucial role in attaching the membrane skeleton to the cell membrane, but they are not exclusive. Considerable experimental data have shown that direct interactions between spectrin and membrane lipids are important for cell membrane cohesion. Spectrin is a multidomain, multifunctional protein with several distinctive structural regions, including lipid-binding sites within CH tandem domains, a PH domain, and triple helical segments, which are excellent examples of ligand specificity hidden in a regular repetitive structure, as recently shown for the ankyrin-sensitive lipid-binding domain of beta spectrin. In this review, we summarize the state of knowledge about interactions between spectrin and membrane lipids.
Collapse
|
45
|
Parry DAD. Fifty years of fibrous protein research: a personal retrospective. J Struct Biol 2013; 186:320-34. [PMID: 24148884 DOI: 10.1016/j.jsb.2013.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 02/02/2023]
Abstract
As a result of X-ray fiber diffraction studies on fibrous proteins and crystallographic data on fragments derived from them, new experimental techniques across the biophysical and biochemical spectra, sophisticated computer modeling and refinement procedures, widespread use of bioinformatics and improved specimen preparative procedures the structures of many fibrous proteins have now been determined to at least low resolution. In so doing these structures have yielded insight into the relationship that exists between sequence and conformation and this, in turn, has led to improved methodologies for predicting structure from sequence data alone. In this personal retrospective a selection of progress made during the past 50years is discussed in terms of events to which the author has made some contribution.
Collapse
Affiliation(s)
- David A D Parry
- Institute of Fundamental Sciences, Massey University, Private Bag 11-222, Palmerston North 4442, New Zealand.
| |
Collapse
|
46
|
Kumeta M, Hirai Y, Yoshimura SH, Horigome T, Takeyasu K. Antibody-based analysis reveals "filamentous vs. non-filamentous" and "cytoplasmic vs. nuclear" crosstalk of cytoskeletal proteins. Exp Cell Res 2013; 319:3226-37. [PMID: 23911988 DOI: 10.1016/j.yexcr.2013.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 06/21/2013] [Accepted: 07/24/2013] [Indexed: 12/01/2022]
Abstract
To uncover the molecular composition and dynamics of the functional scaffold for the nucleus, three fractions of biochemically-stable nuclear protein complexes were extracted and used as immunogens to produce a variety of monoclonal antibodies. Many helix-based cytoskeletal proteins were identified as antigens, suggesting their dynamic contribution to nuclear architecture and function. Interestingly, sets of antibodies distinguished distinct subcellular localization of a single isoform of certain cytoskeletal proteins; distinct molecular forms of keratin and actinin were found in the nucleus. Their nuclear shuttling properties were verified by the apparent nuclear accumulations under inhibition of CRM1-dependent nuclear export. Nuclear keratins do not take an obvious filamentous structure, as was revealed by non-filamentous cytoplasmic keratin-specific monoclonal antibody. These results suggest the distinct roles of the helix-based cytoskeletal proteins in the nucleus.
Collapse
Affiliation(s)
- Masahiro Kumeta
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| | | | | | | | | |
Collapse
|
47
|
Bao J, Bielski E, Bachhawat A, Taha D, Gunther LK, Thirumurugan K, Kitajiri SI, Sakamoto T. R1 motif is the major actin-binding domain of TRIOBP-4. Biochemistry 2013; 52:5256-64. [PMID: 23789641 DOI: 10.1021/bi400585h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
TRIOBP is an actin-bundling protein. Mutations of TRIOBP are associated with human deafness DFNB28. In vitro, TRIOBP isoform 4 (TRIOBP-4) forms dense F-actin bundles resembling the inner ear hair cell rootlet structure. Deletion of TRIOBP isoforms 4 and 5 leads to hearing loss in mice due to the absence of stereocilia rootlets. The mechanism of actin bundle formation by TRIOBP is not fully understood. The amino acid sequences of TRIOBP isoforms 4 and 5 contain two repeated motifs, referred to here as R1 and R2. To examine the potential role of R1 and R2 motifs in F-actin binding, we generated TRIOBP-4 mutant proteins deleted for R1 and/or R2, and then assessed their actin-binding activity and bundle formation in vitro using actin cosedimentation assays, and fluorescence and electron microscopy. Cellular distributions of the TRIOBP-4 mutants were examined by confocal microscopy. We showed that deletion of both R1 and R2 motifs completely disrupted the actin binding/bundling activities of TRIOBP-4 and impaired its localization to cellular actin cytoskeleton structures. By contrast, TRIOBP-4, lacking only R2 motif, retained its F-actin bundling ability and remained localized to actin filaments in cells, similar to full length TRIOBP-4. On the contrary, the R1 motif-deleted TRIOBP-4 mutant, which mainly consists of the R2 motif, formed thin F-actin bundles in vitro but failed to colocalize to actin filaments in cells. These results indicate that R1 motif is the major actin-binding domain of TRIOBP-4, and the binding of R2 motif with actin filaments is nonspecific.
Collapse
Affiliation(s)
- Jianjun Bao
- Department of Physics and Astronomy, Wayne State University , Detroit, Michigan 48201, United States
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Nuclear envelope
spectrin-repeat
proteins (Nesprins), are a novel family of
nuclear and cytoskeletal proteins with rapidly expanding roles as intracellular scaffolds
and linkers. Originally described as proteins that localise to the nuclear envelope (NE)
and establish nuclear-cytoskeletal connections, nesprins have now been found to comprise a
diverse spectrum of tissue specific isoforms that localise to multiple sub-cellular
compartments. Here, we describe how nesprins are necessary in maintaining cellular
architecture by acting as essential scaffolds and linkers at both the NE and other
sub-cellular domains. More importantly, we speculate how nesprin mutations may disrupt
tissue specific nesprin scaffolds and explain the tissue specific nature of many
nesprin-associated diseases, including laminopathies.
Collapse
|
49
|
Machnicka B, Czogalla A, Hryniewicz-Jankowska A, Bogusławska DM, Grochowalska R, Heger E, Sikorski AF. Spectrins: a structural platform for stabilization and activation of membrane channels, receptors and transporters. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:620-34. [PMID: 23673272 DOI: 10.1016/j.bbamem.2013.05.002] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/25/2013] [Accepted: 05/06/2013] [Indexed: 12/22/2022]
Abstract
This review focuses on structure and functions of spectrin as a major component of the membrane skeleton. Recent advances on spectrin function as an interface for signal transduction mediation and a number of data concerning interaction of spectrin with membrane channels, adhesion molecules, receptors and transporters draw a picture of multifaceted protein. Here, we attempted to show the current depiction of multitask role of spectrin in cell physiology. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.
Collapse
Affiliation(s)
- Beata Machnicka
- University of Zielona Góra, Faculty of Biological Sciences, Poland
| | | | | | | | | | - Elżbieta Heger
- University of Zielona Góra, Faculty of Biological Sciences, Poland
| | | |
Collapse
|
50
|
Autore F, Pfuhl M, Quan X, Williams A, Roberts RG, Shanahan CM, Fraternali F. Large-scale modelling of the divergent spectrin repeats in nesprins: giant modular proteins. PLoS One 2013; 8:e63633. [PMID: 23671687 PMCID: PMC3646009 DOI: 10.1371/journal.pone.0063633] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 04/09/2013] [Indexed: 11/29/2022] Open
Abstract
Nesprin-1 and nesprin-2 are nuclear envelope (NE) proteins characterized by a common structure of an SR (spectrin repeat) rod domain and a C-terminal transmembrane KASH [Klarsicht-ANC-Syne-homology] domain and display N-terminal actin-binding CH (calponin homology) domains. Mutations in these proteins have been described in Emery-Dreifuss muscular dystrophy and attributed to disruptions of interactions at the NE with nesprins binding partners, lamin A/C and emerin. Evolutionary analysis of the rod domains of the nesprins has shown that they are almost entirely composed of unbroken SR-like structures. We present a bioinformatical approach to accurate definition of the boundaries of each SR by comparison with canonical SR structures, allowing for a large-scale homology modelling of the 74 nesprin-1 and 56 nesprin-2 SRs. The exposed and evolutionary conserved residues identify important pbs for protein-protein interactions that can guide tailored binding experiments. Most importantly, the bioinformatics analyses and the 3D models have been central to the design of selected constructs for protein expression. 1D NMR and CD spectra have been performed of the expressed SRs, showing a folded, stable, high content α-helical structure, typical of SRs. Molecular Dynamics simulations have been performed to study the structural and elastic properties of consecutive SRs, revealing insights in the mechanical properties adopted by these modules in the cell.
Collapse
Affiliation(s)
- Flavia Autore
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Mark Pfuhl
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
| | - Xueping Quan
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
| | - Aisling Williams
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Roland G. Roberts
- Division of Medical and Molecular Genetics, Kings College London, Guy's Hospital, London, United Kingdom
| | - Catherine M. Shanahan
- Division of Cardiovascular Medicine, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Franca Fraternali
- Randall Division of Cell and Molecular Biophysics, School of Physical Sciences and Engineering, King's College London, London, United Kingdom
- The Thomas Young Centre for Theory and Simulation of Materials, London, United Kingdom
| |
Collapse
|