1
|
Wehn AC, Krestel E, Harapan BN, Klymchenko A, Plesnila N, Khalin I. To see or not to see: In vivo nanocarrier detection methods in the brain and their challenges. J Control Release 2024; 371:216-236. [PMID: 38810705 DOI: 10.1016/j.jconrel.2024.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany.
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Andrey Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin - CS 60024, 67401 Illkirch Cedex, France.
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14 074 Bd Henri Becquerel, 14000 Caen, France.
| |
Collapse
|
2
|
Renart ML, Giudici AM, González-Ros JM, Poveda JA. Steady-state and time-resolved fluorescent methodologies to characterize the conformational landscape of the selectivity filter of K + channels. Methods 2024; 225:89-99. [PMID: 38508347 DOI: 10.1016/j.ymeth.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/02/2024] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
A variety of equilibrium and non-equilibrium methods have been used in a multidisciplinary approach to study the conformational landscape associated with the binding of different cations to the pore of potassium channels. These binding processes, and the conformational changes resulting therefrom, modulate the functional properties of such integral membrane properties, revealing these permeant and blocking cations as true effectors of such integral membrane proteins. KcsA, a prototypic K+ channel from Streptomyces lividans, has been extensively characterized in this regard. Here, we revise several fluorescence-based approaches to monitor cation binding under different experimental conditions in diluted samples, analyzing the advantages and disadvantages of each approach. These studies have contributed to explain the selectivity, conduction, and inactivation properties of K+ channels at the molecular level, together with the allosteric communication between the two gates that control the ion channel flux, and how they are modulated by lipids.
Collapse
Affiliation(s)
- María Lourdes Renart
- IDiBE-Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández, 03202 Elche, Spain.
| | - Ana Marcela Giudici
- IDiBE-Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández, 03202 Elche, Spain.
| | - José M González-Ros
- IDiBE-Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández, 03202 Elche, Spain.
| | - José A Poveda
- IDiBE-Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández, 03202 Elche, Spain.
| |
Collapse
|
3
|
Zhong X, Chen R. Detection of Ferroptosis by Immunohistochemistry and Immunofluorescence. Methods Mol Biol 2023; 2712:211-222. [PMID: 37578709 DOI: 10.1007/978-1-0716-3433-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Ferroptosis is a type of regulated cell death driven by oxidative damage, characterized by iron overload and lipid peroxidation, and regulated by a network of distinct molecules and organelles. Impaired ferroptotic response is implicated in multiple physiological and pathological processes, including tumorigenesis, neurodegeneration, and ischemia-reperfusion damage. Classical techniques of immunohistochemistry (IHC) and immunofluorescence (IF) can be employed to exhibit antigen expression and location in tissues observed with microscopy, making them powerful tools in studying the ferroptosis process. In this chapter, we introduce commonly used protocols and summarize typical markers used in IHC and IF to monitor ferroptosis.
Collapse
Affiliation(s)
- Xiao Zhong
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Characterising ion channel structure and dynamics using fluorescence spectroscopy techniques. Biochem Soc Trans 2022; 50:1427-1445. [DOI: 10.1042/bst20220605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/21/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022]
Abstract
Ion channels undergo major conformational changes that lead to channel opening and ion conductance. Deciphering these structure-function relationships is paramount to understanding channel physiology and pathophysiology. Cryo-electron microscopy, crystallography and computer modelling provide atomic-scale snapshots of channel conformations in non-cellular environments but lack dynamic information that can be linked to functional results. Biophysical techniques such as electrophysiology, on the other hand, provide functional data with no structural information of the processes involved. Fluorescence spectroscopy techniques help bridge this gap in simultaneously obtaining structure-function correlates. These include voltage-clamp fluorometry, Förster resonance energy transfer, ligand binding assays, single molecule fluorescence and their variations. These techniques can be employed to unearth several features of ion channel behaviour. For instance, they provide real time information on local and global rearrangements that are inherent to channel properties. They also lend insights in trafficking, expression, and assembly of ion channels on the membrane surface. These methods have the advantage that they can be carried out in either native or heterologous systems. In this review, we briefly explain the principles of fluorescence and how these have been translated to study ion channel function. We also report several recent advances in fluorescence spectroscopy that has helped address and improve our understanding of the biophysical behaviours of different ion channel families.
Collapse
|
5
|
Li T, Cheng Q, Wang S, Ma C. Rabphilin 3A binds the N-peptide of SNAP-25 to promote SNARE complex assembly in exocytosis. eLife 2022; 11:e79926. [PMID: 36173100 PMCID: PMC9522249 DOI: 10.7554/elife.79926] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Exocytosis of secretory vesicles requires the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins and small GTPase Rabs. As a Rab3/Rab27 effector protein on secretory vesicles, Rabphilin 3A was implicated to interact with SNAP-25 to regulate vesicle exocytosis in neurons and neuroendocrine cells, yet the underlying mechanism remains unclear. In this study, we have characterized the physiologically relevant binding sites between Rabphilin 3A and SNAP-25. We found that an intramolecular interplay between the N-terminal Rab-binding domain and C-terminal C2AB domain enables Rabphilin 3A to strongly bind the SNAP-25 N-peptide region via its C2B bottom α-helix. Disruption of this interaction significantly impaired docking and fusion of vesicles with the plasma membrane in rat PC12 cells. In addition, we found that this interaction allows Rabphilin 3A to accelerate SNARE complex assembly. Furthermore, we revealed that this interaction accelerates SNARE complex assembly via inducing a conformational switch from random coils to α-helical structure in the SNAP-25 SNARE motif. Altogether, our data suggest that the promotion of SNARE complex assembly by binding the C2B bottom α-helix of Rabphilin 3A to the N-peptide of SNAP-25 underlies a pre-fusion function of Rabphilin 3A in vesicle exocytosis.
Collapse
Affiliation(s)
- Tianzhi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Qiqi Cheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
6
|
Arias A, Manubens-Gil L, Dierssen M. Fluorescent transgenic mouse models for whole-brain imaging in health and disease. Front Mol Neurosci 2022; 15:958222. [PMID: 36211979 PMCID: PMC9538927 DOI: 10.3389/fnmol.2022.958222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
A paradigm shift is occurring in neuroscience and in general in life sciences converting biomedical research from a descriptive discipline into a quantitative, predictive, actionable science. Living systems are becoming amenable to quantitative description, with profound consequences for our ability to predict biological phenomena. New experimental tools such as tissue clearing, whole-brain imaging, and genetic engineering technologies have opened the opportunity to embrace this new paradigm, allowing to extract anatomical features such as cell number, their full morphology, and even their structural connectivity. These tools will also allow the exploration of new features such as their geometrical arrangement, within and across brain regions. This would be especially important to better characterize brain function and pathological alterations in neurological, neurodevelopmental, and neurodegenerative disorders. New animal models for mapping fluorescent protein-expressing neurons and axon pathways in adult mice are key to this aim. As a result of both developments, relevant cell populations with endogenous fluorescence signals can be comprehensively and quantitatively mapped to whole-brain images acquired at submicron resolution. However, they present intrinsic limitations: weak fluorescent signals, unequal signal strength across the same cell type, lack of specificity of fluorescent labels, overlapping signals in cell types with dense labeling, or undetectable signal at distal parts of the neurons, among others. In this review, we discuss the recent advances in the development of fluorescent transgenic mouse models that overcome to some extent the technical and conceptual limitations and tradeoffs between different strategies. We also discuss the potential use of these strains for understanding disease.
Collapse
Affiliation(s)
- Adrian Arias
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Linus Manubens-Gil
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Mara Dierssen
- Department of System Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Department of Experimental and Health Sciences, University Pompeu Fabra, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
7
|
Alhazmi S, Alzahrani M, Farsi R, Alharbi M, Algothmi K, Alburae N, Ganash M, Azhari S, Basingab F, Almuhammadi A, Alqosaibi A, Alkhatabi H, Elaimi A, Jan M, Aldhalaan HM, Alrafiah A, Alrofaidi A. Multiple Recurrent Copy Number Variations (CNVs) in Chromosome 22 Including 22q11.2 Associated with Autism Spectrum Disorder. Pharmgenomics Pers Med 2022; 15:705-720. [PMID: 35898556 PMCID: PMC9309317 DOI: 10.2147/pgpm.s366826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/14/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Autism spectrum disorder (ASD) is a developmental disorder that can cause substantial social, communication, and behavioral challenges. Genetic factors play a significant role in ASD, where the risk of ASD has been increased for unclear reasons. Twin studies have shown important evidence of both genetic and environmental contributions in ASD, where the level of contribution of these factors has not been proven yet. It has been suggested that copy number variation (CNV) duplication and the deletion of many genes in chromosome 22 (Ch22) may have a strong association with ASD. This study screened the CNVs in Ch22 in autistic Saudi children and assessed the candidate gene in the CNVs region of Ch22 that is most associated with ASD. Methods This study included 15 autistic Saudi children as well as 4 healthy children as controls; DNA was extracted from samples and analyzed using array comparative genomic hybridization (aCGH) and DNA sequencing. Results The aCGH detected (in only 6 autistic samples) deletion and duplication in many regions of Ch22, including some critical genes. Moreover, DNA sequencing determined a genetic mutation in the TBX1 gene sequence in autistic samples. This study, carried out using aCGH, found that six autistic patients had CNVs in Ch22, and DNA sequencing revealed mutations in the TBX1 gene in autistic samples but none in the control. Conclusion CNV deletion and the duplication of the TBX1 gene could be related to ASD; therefore, this gene needs more analysis in terms of expression levels.
Collapse
Affiliation(s)
- Safiah Alhazmi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maryam Alzahrani
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Reem Farsi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Alharbi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Khloud Algothmi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Najla Alburae
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Magdah Ganash
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sheren Azhari
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Asma Almuhammadi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amany Alqosaibi
- Department of Biology, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Heba Alkhatabi
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Aisha Elaimi
- Centre of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Jan
- College of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hesham M Aldhalaan
- Center for Autism Research at King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Aziza Alrafiah
- Department of Medical Laboratory Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Correspondence: Aziza Alrafiah, Department of Medical Laboratory Science, King Abdulaziz University, P.O Box 80200, Jeddah, 21589, Saudi Arabia, Tel +966 126401000 Ext. 23495, Fax +966 126401000 Ext. 21686, Email
| | - Aisha Alrofaidi
- Department of Biological Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Pham ATT, Tohl D, Wallace A, Hu Q, Li J, Reynolds KJ, Tang Y. Developing a fluorescent sensing based portable medical open-platform - a case study for albuminuria measurement in chronic kidney disease screening and monitoring. SENSING AND BIO-SENSING RESEARCH 2022. [DOI: 10.1016/j.sbsr.2022.100504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
9
|
Bajer D, Kaczmarek H. Thermal Stability of Fluorescent Chitosan Modified with Heterocyclic Aromatic Dyes. MATERIALS 2022; 15:ma15103667. [PMID: 35629691 PMCID: PMC9147818 DOI: 10.3390/ma15103667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/07/2022]
Abstract
Fluorescent biopolymer derivatives are increasingly used in biology and medicine, but their resistance to heat and UV radiation, which are sterilizing agents, is relatively unknown. In this work, chitosan (CS) modified by three different heterocyclic aromatic dyes based on benzimidazole, benzothiazole, and benzoxazole (assigned as IBm, BTh, and BOx) has been studied. The thermal properties of these CS derivatives have been determined using the Thermogravimetric Analysis coupled with the Fourier Transform Infrared spectroscopy of volatile degradation products. The influence of UV radiation on the thermal resistance of modified, fluorescent chitosan samples was also investigated. Based on the temperature onset as well as the decomposition temperatures at a maximal rate, IBm was found to be more thermally stable than BOx and BTh. However, this dye gave off the most volatile products (mainly water, ammonia, carbon oxides, and carbonyl/ether compounds). The substitution of dyes for chitosan changes its thermal stability slightly. Characteristic decomposition temperatures in modified CS vary by a few degrees (<10 °C) from the virgin sample. Considering the temperatures of the main decomposition stage, CS-BOx turned out to be the most stable. The UV irradiation of chitosan derivatives leads to minor changes in the thermal parameters and a decrease in the number of volatile degradation products. It was concluded that the obtained CS derivatives are characterized by good resistance to heat and UV irradiation, which extends the possibilities of using these innovative materials.
Collapse
Affiliation(s)
- Dagmara Bajer
- Correspondence: (D.B.); (H.K.); Tel.: +48-56-611-4505 (D.B.); +48-56-611-4312 (H.K.)
| | - Halina Kaczmarek
- Correspondence: (D.B.); (H.K.); Tel.: +48-56-611-4505 (D.B.); +48-56-611-4312 (H.K.)
| |
Collapse
|
10
|
Kelmanson IV, Shokhina AG, Kotova DA, Pochechuev MS, Ivanova AD, Kostyuk AI, Panova AS, Borodinova AA, Solotenkov MA, Stepanov EA, Raevskii RI, Moshchenko AA, Pak VV, Ermakova YG, van Belle GJC, Tarabykin V, Balaban PM, Fedotov IV, Fedotov AB, Conrad M, Bogeski I, Katschinski DM, Doeppner TR, Bähr M, Zheltikov AM, Belousov VV, Bilan DS. In vivo dynamics of acidosis and oxidative stress in the acute phase of an ischemic stroke in a rodent model. Redox Biol 2021; 48:102178. [PMID: 34773835 PMCID: PMC8600061 DOI: 10.1016/j.redox.2021.102178] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 02/08/2023] Open
Abstract
Ischemic cerebral stroke is one of the leading causes of death and disability in humans. However, molecular processes underlying the development of this pathology remain poorly understood. There are major gaps in our understanding of metabolic changes that occur in the brain tissue during the early stages of ischemia and reperfusion. In particular, it is generally accepted that both ischemia (I) and reperfusion (R) generate reactive oxygen species (ROS) that cause oxidative stress which is one of the main drivers of the pathology, although ROS generation during I/R was never demonstrated in vivo due to the lack of suitable methods. In the present study, we record for the first time the dynamics of intracellular pH and H2O2 during I/R in cultured neurons and during experimental stroke in rats using the latest generation of genetically encoded biosensors SypHer3s and HyPer7. We detect a buildup of powerful acidosis in the brain tissue that overlaps with the ischemic core from the first seconds of pathogenesis. At the same time, no significant H2O2 generation was found in the acute phase of ischemia/reperfusion. HyPer7 oxidation in the brain was detected only 24 h later. Comparison of in vivo experiments with studies on cultured neurons under I/R demonstrates that the dynamics of metabolic processes in these models significantly differ, suggesting that a cell culture is a poor predictor of metabolic events in vivo.
Collapse
Affiliation(s)
- Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Arina G Shokhina
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia
| | - Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Matvei S Pochechuev
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Alexandra D Ivanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Biological Department, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Alexander I Kostyuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Anastasiya S Panova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Anastasia A Borodinova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Maxim A Solotenkov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia
| | - Evgeny A Stepanov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia
| | - Roman I Raevskii
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Aleksandr A Moshchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia
| | - Valeriy V Pak
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Yulia G Ermakova
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | - Gijsbert J C van Belle
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Viktor Tarabykin
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Pavel M Balaban
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, 117485, Russia
| | - Ilya V Fedotov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia; Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, 420126, Russia; Department of Physics and Astronomy, Texas A&M University, College Station, TX, 77843, USA
| | - Andrei B Fedotov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia
| | - Marcus Conrad
- Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany
| | - Ivan Bogeski
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Dörthe M Katschinski
- Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, 37075, Germany; Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Istanbul, Turkey; Istanbul Medipol University, School of Medicine, Dept. of Physiology, Istanbul, Turkey
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, 37075, Germany
| | - Aleksei M Zheltikov
- Physics Department, International Laser Center, M.V. Lomonosov Moscow State University, Moscow, 119992, Russia; Russian Quantum Center, Skolkovo, Moscow Region, 143025, Russia; Kazan Quantum Center, A.N. Tupolev Kazan National Research Technical University, Kazan, 420126, Russia; Department of Physics and Astronomy, Texas A&M University, College Station, TX, 77843, USA
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117997, Russia; Institute for Cardiovascular Physiology, University Medical Center Göttingen, Georg-August-University, Humboldtallee 23, 37073, Göttingen, Germany.
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia; Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| |
Collapse
|
11
|
Jana R, Begam HM, Dinda E. The emergence of the C-H functionalization strategy in medicinal chemistry and drug discovery. Chem Commun (Camb) 2021; 57:10842-10866. [PMID: 34596175 DOI: 10.1039/d1cc04083a] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Owing to the market competitiveness and urgent societal need, an optimum speed of drug discovery is an important criterion for successful implementation. Despite the rapid ascent of artificial intelligence and computational and bioanalytical techniques to accelerate drug discovery in big pharma, organic synthesis of privileged scaffolds predicted in silico for in vitro and in vivo studies is still considered as the rate-limiting step. C-H activation is the latest technology added into an organic chemist's toolbox for the rapid construction and late-stage modification of functional molecules to achieve the desired chemical and physical properties. Particularly, elimination of prefunctionalization steps, exceptional functional group tolerance, complexity-to-diversity oriented synthesis, and late-stage functionalization of privileged medicinal scaffolds expand the chemical space. It has immense potential for the rapid synthesis of a library of molecules, structural modification to achieve the required pharmacological properties such as absorption, distribution, metabolism, excretion, toxicology (ADMET) and attachment of chemical reporters for proteome profiling, metabolite synthesis, etc. for preclinical studies. Although heterocycle synthesis, late-stage drug modification, 18F labelling, methylation, etc. via C-H functionalization have been reviewed from the synthetic standpoint, a general overview of these protocols from medicinal and drug discovery aspects has not been reviewed. In this feature article, we will discuss the recent trends of C-H activation methodologies such as synthesis of medicinal scaffolds through C-H activation/annulation cascade; C-H arylation for sp2-sp2 and sp2-sp3 cross-coupling; C-H borylation/silylation to introduce a functional linchpin for further manipulation; C-H amination for N-heterocycles and hydrogen bond acceptors; C-H fluorination/fluoroalkylation to tune polarity and lipophilicity; C-H methylation: methyl magic in drug discovery; peptide modification and macrocyclization for therapeutics and biologics; fluorescent labelling and radiolabelling for bioimaging; bioconjugation for chemical biology studies; drug-metabolite synthesis for biodistribution and excretion studies; late-stage diversification of drug-molecules to increase efficacy and safety; cutting-edge DNA encoded library synthesis and improved synthesis of drug molecules via C-H activation in medicinal chemistry and drug discovery.
Collapse
Affiliation(s)
- Ranjan Jana
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata-700032, India.
| | - Hasina Mamataj Begam
- Organic and Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata-700032, India.
| | - Enakshi Dinda
- Department of Chemistry and Environment, Heritage Institute of Technology, Kolkata-700107, India
| |
Collapse
|
12
|
Pham ATT, Wallace A, Zhang X, Tohl D, Fu H, Chuah C, Reynolds KJ, Ramsey C, Tang Y. Optical-Based Biosensors and Their Portable Healthcare Devices for Detecting and Monitoring Biomarkers in Body Fluids. Diagnostics (Basel) 2021; 11:diagnostics11071285. [PMID: 34359368 PMCID: PMC8307945 DOI: 10.3390/diagnostics11071285] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
The detection and monitoring of biomarkers in body fluids has been used to improve human healthcare activities for decades. In recent years, researchers have focused their attention on applying the point-of-care (POC) strategies into biomarker detection. The evolution of mobile technologies has allowed researchers to develop numerous portable medical devices that aim to deliver comparable results to clinical measurements. Among these, optical-based detection methods have been considered as one of the common and efficient ways to detect and monitor the presence of biomarkers in bodily fluids, and emerging aggregation-induced emission luminogens (AIEgens) with their distinct features are merging with portable medical devices. In this review, the detection methodologies that use optical measurements in the POC systems for the detection and monitoring of biomarkers in bodily fluids are compared, including colorimetry, fluorescence and chemiluminescence measurements. The current portable technologies, with or without the use of smartphones in device development, that are combined with optical biosensors for the detection and monitoring of biomarkers in body fluids, are also investigated. The review also discusses novel AIEgens used in the portable systems for the detection and monitoring of biomarkers in body fluid. Finally, the potential of future developments and the use of optical detection-based portable devices in healthcare activities are explored.
Collapse
Affiliation(s)
- Anh Tran Tam Pham
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Angus Wallace
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Xinyi Zhang
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Damian Tohl
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Hao Fu
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Clarence Chuah
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Karen J. Reynolds
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Carolyn Ramsey
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
| | - Youhong Tang
- Australia-China Science and Research Fund Joint Research Centre for Personal Health Technologies, Flinders University, Tonsley, SA 5042, Australia; (A.T.T.P.); (A.W.); (X.Z.); (D.T.); (H.F.); (K.J.R.); (C.R.)
- Medical Device Research Institute, Flinders University, Tonsley, SA 5042, Australia;
- Correspondence: ; Tel.: +61-8-8201-2138
| |
Collapse
|
13
|
Electromechanical coupling mechanism for activation and inactivation of an HCN channel. Nat Commun 2021; 12:2802. [PMID: 33990563 PMCID: PMC8121817 DOI: 10.1038/s41467-021-23062-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/01/2021] [Indexed: 11/08/2022] Open
Abstract
Pacemaker hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels exhibit a reversed voltage-dependent gating, activating by membrane hyperpolarization instead of depolarization. Sea urchin HCN (spHCN) channels also undergo inactivation with hyperpolarization which occurs only in the absence of cyclic nucleotide. Here we applied transition metal ion FRET, patch-clamp fluorometry and Rosetta modeling to measure differences in the structural rearrangements between activation and inactivation of spHCN channels. We found that removing cAMP produced a largely rigid-body rotation of the C-linker relative to the transmembrane domain, bringing the A’ helix of the C-linker in close proximity to the voltage-sensing S4 helix. In addition, rotation of the C-linker was elicited by hyperpolarization in the absence but not the presence of cAMP. These results suggest that — in contrast to electromechanical coupling for channel activation — the A’ helix serves to couple the S4-helix movement for channel inactivation, which is likely a conserved mechanism for CNBD-family channels. Sea urchin hyperpolarization-activated cyclic nucleotide-gated (spHCN) ion channels channels are activated by membrane hyperpolarization instead of depolarization and undergo inactivation with hyperpolarization. Here authors apply transition metal ion FRET, patch-clamp fluorometry and Rosetta modeling to measure differences in the structural rearrangements between activation and inactivation of spHCN channels.
Collapse
|
14
|
Rocha MA, Sprague-Piercy MA, Kwok AO, Roskamp KW, Martin RW. Chemical Properties Determine Solubility and Stability in βγ-Crystallins of the Eye Lens. Chembiochem 2021; 22:1329-1346. [PMID: 33569867 PMCID: PMC8052307 DOI: 10.1002/cbic.202000739] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/17/2020] [Indexed: 11/10/2022]
Abstract
βγ-Crystallins are the primary structural and refractive proteins found in the vertebrate eye lens. Because crystallins are not replaced after early eye development, their solubility and stability must be maintained for a lifetime, which is even more remarkable given the high protein concentration in the lens. Aggregation of crystallins caused by mutations or post-translational modifications can reduce crystallin protein stability and alter intermolecular interactions. Common post-translational modifications that can cause age-related cataracts include deamidation, oxidation, and tryptophan derivatization. Metal ion binding can also trigger reduced crystallin solubility through a variety of mechanisms. Interprotein interactions are critical to maintaining lens transparency: crystallins can undergo domain swapping, disulfide bonding, and liquid-liquid phase separation, all of which can cause opacity depending on the context. Important experimental techniques for assessing crystallin conformation in the absence of a high-resolution structure include dye-binding assays, circular dichroism, fluorescence, light scattering, and transition metal FRET.
Collapse
Affiliation(s)
- Megan A. Rocha
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
| | - Marc A. Sprague-Piercy
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697-2525
| | - Ashley O. Kwok
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
| | - Kyle W. Roskamp
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
| | - Rachel W. Martin
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences 2, Irvine, CA 92697-2025 (USA)
- Department of Molecular Biology and Biochemistry, University of California Irvine, 3205 McGaugh Hall, Irvine, CA 92697-2525
| |
Collapse
|
15
|
Marvian AT, Aliakbari F, Mohammad-Beigi H, Ahmadi ZA, Mehrpooyan S, Lermyte F, Nasouti M, Collingwood JF, Otzen DE, Morshedi D. The status of the terminal regions of α-synuclein in different forms of aggregates during fibrillization. Int J Biol Macromol 2020; 155:543-550. [PMID: 32240735 DOI: 10.1016/j.ijbiomac.2020.03.238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 10/24/2022]
Abstract
The α-synuclein (αSN) amyloid fibrillization process is known to be a crucial phenomenon associated with neuronal loss in various neurodegenerative diseases, most famously Parkinson's disease. The process involves different aggregated species and ultimately leads to formation of β-sheet rich fibrillar structures. Despite the essential role of αSN aggregation in the pathoetiology of various neurological disorders, the characteristics of various assemblies are not fully understood. Here, we established a fluorescence-based model for studying the end-parts of αSN to decipher the structural aspects of aggregates during the fibrillization. Our model proved highly sensitive to the events at the early stage of the fibrillization process, which are hardly detectable with routine techniques. Combining fluorescent and PAGE analysis, we found different oligomeric aggregates in the nucleation phase of fibrillization with different sensitivity to SDS and different structures based on αSN termini. Moreover, we found that these oligomers are highly dynamic: after reaching peak levels during fibrillization, they decline and eventually disappear, suggesting their transformation into other αSN aggregated species. These findings shed light on the structural features of various αSN aggregates and their dynamics in synucleinopathies.
Collapse
Affiliation(s)
- Amir Tayaranian Marvian
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany; Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Farhang Aliakbari
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran; Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Hossein Mohammad-Beigi
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Zeinab Alsadat Ahmadi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Sina Mehrpooyan
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Mahour Nasouti
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Daniel E Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Dina Morshedi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
16
|
Mahmoud ME, Moussa Z, Prakasam T, Li L, Abiad MG, Patra D, Hmadeh M. Lanthanides based metal organic frameworks for luminescence sensing of toxic metal ions. J SOLID STATE CHEM 2020. [DOI: 10.1016/j.jssc.2019.121031] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
17
|
Gupta K, Toombes GE, Swartz KJ. Exploring structural dynamics of a membrane protein by combining bioorthogonal chemistry and cysteine mutagenesis. eLife 2019; 8:50776. [PMID: 31714877 PMCID: PMC6850778 DOI: 10.7554/elife.50776] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The functional mechanisms of membrane proteins are extensively investigated with cysteine mutagenesis. To complement cysteine-based approaches, we engineered a membrane protein with thiol-independent crosslinkable groups using azidohomoalanine (AHA), a non-canonical methionine analogue containing an azide group that can selectively react with cycloalkynes through a strain-promoted azide-alkyne cycloaddition (SPAAC) reaction. We demonstrate that AHA can be readily incorporated into the Shaker Kv channel in place of methionine residues and modified with azide-reactive alkyne probes in Xenopus oocytes. Using voltage-clamp fluorometry, we show that AHA incorporation permits site-specific fluorescent labeling to track voltage-dependent conformational changes similar to cysteine-based methods. By combining AHA incorporation and cysteine mutagenesis in an orthogonal manner, we were able to site-specifically label the Shaker Kv channel with two different fluorophores simultaneously. Our results identify a facile and straightforward approach for chemical modification of membrane proteins with bioorthogonal chemistry to explore their structure-function relationships in live cells. Living cells can sense cues from their environment via molecules located at the interface between the inside and the outside of the cell. These molecules are mostly proteins and are made up of building blocks known as amino acids. To understand how these proteins work, fluorescent probes can be attached to amino acids within them – which can then tell when different parts of proteins move in response to a signal. Scientists often target fluorescent probes at the amino acid cysteine, because it has a chemically reactive side group and is rare enough so that unique positions can be labeled in the protein of interest. However, being able to target other amino acids would allow scientists to ask, and potentially solve, more precise questions about these proteins. Methionine is another amino acid that has a low abundance in most proteins. Previous research has shown that the cell’s normal protein-building machinery can incorporate synthetic versions of methionine into proteins. This suggested that the introduction of chemically reactive alternatives to methionine could offer a way to label membrane proteins with fluorescent probes and free up the cysteines to be targeted with other approaches. Gupta et al. set out to develop a straightforward method to achieve this and started with a well-studied membrane protein, called Shaker, and cells from female African clawed frogs, which are widely used to study membrane proteins. Gupta et al. found that the cells could readily take up a chemically reactive methionine alternative called azidohomoalanine (AHA) from their surrounding solution and incorporate it within the Shaker protein. The AHA took the place of the methionines that are normally found in Shaker, and just like in cysteine-based methods, fluorescent probes could be easily attached to the AHAs in this membrane protein. Shaker is a protein that allows potassium ions to flow across the cell membrane by changing shape in response to the membrane voltage. The fluorescence from those probes also changed with the membrane voltage in a way that was comparable to cysteine-mediated approaches. This indicated that the AHA modification could also be used to track structural changes in the Shaker protein. Finally, Gupta et al. showed that AHA- and cysteine-mediated labeling approaches could be combined to attach two different fluorescent probes onto the Shaker protein. This method will expand the toolbox for researchers studying the relationship between the structure and function of membrane proteins in live cells. In future, it could be applied more widely once the properties of the fluorescent probes for AHA-mediated labeling can be optimized.
Collapse
Affiliation(s)
- Kanchan Gupta
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, United States
| | - Gilman Es Toombes
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, United States
| | - Kenton J Swartz
- Molecular Physiology and Biophysics Section, Porter Neuroscience Research Center, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|
18
|
Zhang DY, Singhal S, Lee JYK. Optical Principles of Fluorescence-Guided Brain Tumor Surgery: A Practical Primer for the Neurosurgeon. Neurosurgery 2019; 85:312-324. [PMID: 30085129 DOI: 10.1093/neuros/nyy315] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 06/18/2018] [Indexed: 01/21/2023] Open
Abstract
Fluorescence-guided surgery is a rapidly growing field that has produced some of the most important innovations in surgical oncology in the past decade. These intraoperative imaging technologies provide information distinguishing tumor tissue from normal tissue in real time as the surgery proceeds and without disruption of the workflow. Many of these fluorescent tracers target unique molecular or cellular features of tumors, which offers the opportunity for identifying pathology with high precision to help surgeons achieve their primary objective of a maximal safe resection. As novel fluorophores and fluorescent probes emerge from preclinical development, a practical understanding of the principles of fluorescence remains critical for evaluating the clinical utility of these agents and identifying opportunities for further innovation. In this review, we provide an "in-text glossary" of the fundamental principles of fluorescence with examples of direct applications to fluorescence-guided brain surgery. We offer a detailed discussion of the various advantages and limitations of the most commonly used intraoperative imaging agents, including 5-aminolevulinic acid, indocyanine green, and fluorescein, with a particular focus on the photophysical properties of these specific agents as they provide a framework through which to understand the new agents that are entering clinical trials. To this end, we conclude with a survey of the fluorescent properties of novel agents that are currently undergoing or will soon enter clinical trials for the intraoperative imaging of brain tumors.
Collapse
Affiliation(s)
- Daniel Y Zhang
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Y K Lee
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Taraska JW. A primer on resolving the nanoscale structure of the plasma membrane with light and electron microscopy. J Gen Physiol 2019; 151:974-985. [PMID: 31253697 PMCID: PMC6683668 DOI: 10.1085/jgp.201812227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/10/2019] [Indexed: 12/20/2022] Open
Abstract
Taraska reviews the imaging methods that are being used to understand the structure of the plasma membrane at the molecular level. The plasma membrane separates a cell from its external environment. All materials and signals that enter or leave the cell must cross this hydrophobic barrier. Understanding the architecture and dynamics of the plasma membrane has been a central focus of general cellular physiology. Both light and electron microscopy have been fundamental in this endeavor and have been used to reveal the dense, complex, and dynamic nanoscale landscape of the plasma membrane. Here, I review classic and recent developments in the methods used to image and study the structure of the plasma membrane, particularly light, electron, and correlative microscopies. I will discuss their history and use for mapping the plasma membrane and focus on how these tools have provided a structural framework for understanding the membrane at the scale of molecules. Finally, I will describe how these studies provide a roadmap for determining the nanoscale architecture of other organelles and entire cells in order to bridge the gap between cellular form and function.
Collapse
Affiliation(s)
- Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
20
|
Shi H, Ma S, Zhang R, Yuan J. A hidden state in the turnover of a functioning membrane protein complex. SCIENCE ADVANCES 2019; 5:eaau6885. [PMID: 30906857 PMCID: PMC6426456 DOI: 10.1126/sciadv.aau6885] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/30/2019] [Indexed: 06/09/2023]
Abstract
Most membrane proteins exist in complexes that carry out critical cellular functions and exhibit rich dynamics. The bacterial flagellar motor, a large membrane-spanning ion-driven rotary motor that propels the bacteria to swim, provides a canonical example. Rotation of the motor is driven by multiple torque-generating units (stators). Turnover of the stators has been shown previously, demonstrating the exchange of stator units between the motor and a membrane pool. But the details of the turnover kinetics remain unclear. Here, we directly measured the kinetics of stator turnover in individual motors via analysis of a large dataset of long-term high-resolution recordings of motor speed at high load. We found that the dwell time distribution of the stator units exhibits a multi-exponential shape, suggesting the existence of a hidden state in the turnover of the stators.
Collapse
|
21
|
Gordon SE, Munari M, Zagotta WN. Visualizing conformational dynamics of proteins in solution and at the cell membrane. eLife 2018; 7:37248. [PMID: 29923827 PMCID: PMC6056233 DOI: 10.7554/elife.37248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/05/2018] [Indexed: 01/03/2023] Open
Abstract
Conformational dynamics underlie enzyme function, yet are generally inaccessible via traditional structural approaches. FRET has the potential to measure conformational dynamics in vitro and in intact cells, but technical barriers have thus far limited its accuracy, particularly in membrane proteins. Here, we combine amber codon suppression to introduce a donor fluorescent noncanonical amino acid with a new, biocompatible approach for labeling proteins with acceptor transition metals in a method called ACCuRET (Anap Cyclen-Cu2+ resonance energy transfer). We show that ACCuRET measures absolute distances and distance changes with high precision and accuracy using maltose binding protein as a benchmark. Using cell unroofing, we show that ACCuRET can accurately measure rearrangements of proteins in native membranes. Finally, we implement a computational method for correcting the measured distances for the distance distributions observed in proteins. ACCuRET thus provides a flexible, powerful method for measuring conformational dynamics in both soluble proteins and membrane proteins.
Collapse
Affiliation(s)
- Sharona E Gordon
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - Mika Munari
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, United States
| |
Collapse
|
22
|
Manzoor O, Soleja N, Mohsin M. Nanoscale gizmos - the novel fluorescent probes for monitoring protein activity. Biochem Eng J 2018; 133:83-95. [PMID: 32518506 PMCID: PMC7270366 DOI: 10.1016/j.bej.2018.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/27/2017] [Accepted: 02/06/2018] [Indexed: 11/15/2022]
Abstract
Genetically-encoded FRET, organic dye, QD based sensors. Real-time monitoring of the respective metabolite level at sub cellular level. Spatio temporal resolution of the fluorophores by low intensity light. Monitoring of various metabolite levels in any cell type prokaryotic and eukaryotic as well. Functional analysis of the role of proteases in several diseases.
Nanobiotechnology has emerged inherently as an interdisciplinary field, with collaborations from researchers belonging to diverse backgrounds like molecular biology, materials science and organic chemistry. Till the current times, researchers have been able to design numerous types of nanoscale fluorescent tool kits for monitoring protein–protein interactions through real time cellular imagery in a fluorescence microscope. It is apparent that supplementing any protein of interest with a fluorescence habit traces its function and regulation within a cell. Our review therefore highlights the application of several fluorescent probes such as molecular organic dyes, quantum dots (QD) and fluorescent proteins (FPs) to determine activity state, expression and localization of proteins in live and fixed cells. The focus is on Fluorescence Resonance Energy Transfer (FRET) based nanosensors that have been developed by researchers to visualize and monitor protein dynamics and quantify metabolites of diverse nature. FRET based toolkits permit the resolution of ambiguities that arise due to the rotation of sensor molecules and flexibility of the probe. Achievements of live cell imaging and efficient spatiotemporal resolution however have been possible only with the advent of fluorescence microscopic technology, equipped with precisely sensitive automated softwares.
Collapse
|
23
|
Fabilane CS, Nguyen PN, Hernandez RV, Nirudodhi S, Duong M, Maier CS, Narayanaswami V. Mechanism of Lipid Binding of Human Apolipoprotein E3 by Hydrogen/Deuterium Exchange/Mass Spectrometry and Fluorescence Polarization. Protein Pept Lett 2016; 23:404-13. [PMID: 26902251 DOI: 10.2174/0929866523666160223122257] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 11/22/2022]
Abstract
BACKGROUND Human apolipoprotein E3 (apoE3) is an exchangeable apolipoprotein that plays a critical role in maintaining plasma cholesterol/triglyceride homeostasis. The C-terminal (CT) domain of apoE3 (residues 201-299) is composed of amphipathic α-helices C1: W210-S223, C2: V236-E266, and C3: D271-W276, which play a dominant role in mediating high-affinity lipid binding. OBJECTIVE The objective is to understand the accessibility of the CT domain at the sub-domain level and the mechanistic details regarding lipid-binding interaction. METHODS Hydrogen-deuterium exchange coupled to mass spectrometry (HDX/MS) of recombinant wild type (WT) apoE(201-299), chemical-induced unfolding monitored as changes in fluorescence polarization (FP) of labeled apoE(201-299) bearing a probe at specified sites, and lipid binding studies were carried out. RESULTS HDX/MS revealed that residues towards the C-terminal end of the domain display significantly lower %D uptake compared to those towards the center, suggesting extensive protein-protein interaction in this segment. Functional assays showed that locking apoE(201-299) in an inter-molecular disulfide-bonded state at position 209, 223, 255, or 277 significantly decreases its ability to interact with lipids, especially when tethered towards the ends; this could be restored by reduction. Unfolding studies indicate that the C-terminal end offers less resistance to unfolding compared to the central portion of the domain. CONCLUSION Taken together, our data suggest that two dimers of CT domain are juxtaposed around helix C3 leading to apoE3 tetramerization, and that dissociation to monomeric units is a required step in lipid binding, with helix C3 likely seeking stability via lipid interaction prior to helices C1 or C2.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vasanthy Narayanaswami
- Department of Chemistry & Biochemistry, 1250 Bellflower Blvd, California State University Long Beach, Long Beach, CA 90840, USA.
| |
Collapse
|
24
|
Ozgen H, Baron W, Hoekstra D, Kahya N. Oligodendroglial membrane dynamics in relation to myelin biogenesis. Cell Mol Life Sci 2016; 73:3291-310. [PMID: 27141942 PMCID: PMC4967101 DOI: 10.1007/s00018-016-2228-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/14/2016] [Indexed: 12/12/2022]
Abstract
In the central nervous system, oligodendrocytes synthesize a specialized membrane, the myelin membrane, which enwraps the axons in a multilamellar fashion to provide fast action potential conduction and to ensure axonal integrity. When compared to other membranes, the composition of myelin membranes is unique with its relatively high lipid to protein ratio. Their biogenesis is quite complex and requires a tight regulation of sequential events, which are deregulated in demyelinating diseases such as multiple sclerosis. To devise strategies for remedying such defects, it is crucial to understand molecular mechanisms that underlie myelin assembly and dynamics, including the ability of specific lipids to organize proteins and/or mediate protein-protein interactions in healthy versus diseased myelin membranes. The tight regulation of myelin membrane formation has been widely investigated with classical biochemical and cell biological techniques, both in vitro and in vivo. However, our knowledge about myelin membrane dynamics, such as membrane fluidity in conjunction with the movement/diffusion of proteins and lipids in the membrane and the specificity and role of distinct lipid-protein and protein-protein interactions, is limited. Here, we provide an overview of recent findings about the myelin structure in terms of myelin lipids, proteins and membrane microdomains. To give insight into myelin membrane dynamics, we will particularly highlight the application of model membranes and advanced biophysical techniques, i.e., approaches which clearly provide an added value to insight obtained by classical biochemical techniques.
Collapse
Affiliation(s)
- Hande Ozgen
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Wia Baron
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | - Dick Hoekstra
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Nicoletta Kahya
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
25
|
Finkenstaedt-Quinn SA, Qiu TA, Shin K, Haynes CL. Super-resolution imaging for monitoring cytoskeleton dynamics. Analyst 2016; 141:5674-5688. [PMID: 27549146 DOI: 10.1039/c6an00731g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cytoskeleton is a key cellular structure that is important in the control of cellular movement, structure, and sensing. To successfully image the individual cytoskeleton components, high resolution and super-resolution fluorescence imaging methods are needed. This review covers the three basic cytoskeletal elements and the relative benefits and drawbacks of fixed versus live cell imaging before moving on to recent studies using high resolution and super-resolution techniques. The techniques covered include the near-diffraction limited imaging methods of confocal microscopy and TIRF microscopy and the super-resolution fluorescence imaging methods of STORM, PALM, and STED.
Collapse
|
26
|
Wang S, Li Y, Ma C. Synaptotagmin-1 C2B domain interacts simultaneously with SNAREs and membranes to promote membrane fusion. eLife 2016; 5. [PMID: 27083046 PMCID: PMC4878868 DOI: 10.7554/elife.14211] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/14/2016] [Indexed: 12/30/2022] Open
Abstract
Synaptotagmin-1 (Syt1) acts as a Ca2+ sensor for neurotransmitter release through its C2 domains. It has been proposed that Syt1 promotes SNARE-dependent fusion mainly through its C2B domain, but the underlying mechanism is poorly understood. In this study, we show that the C2B domain interacts simultaneously with acidic membranes and SNARE complexes via the top Ca2+-binding loops, the side polybasic patch, and the bottom face in response to Ca2+. Disruption of the simultaneous interactions completely abrogates the triggering activity of the C2B domain in liposome fusion. We hypothesize that the simultaneous interactions endow the C2B domain with an ability to deform local membranes, and this membrane-deformation activity might underlie the functional significance of the Syt1 C2B domain in vivo. DOI:http://dx.doi.org/10.7554/eLife.14211.001 Information travels around the nervous system along cells called neurons, which communicate with each other via connections called synapses. When a signal travelling along one neuron reaches a synapse, it triggers the release of molecules known as neurotransmitters. These molecules are then taken up by the next neuron to pass the signal on. Neurotransmitters are stored in compartments called synaptic vesicles and their release from the first neuron depends on the synaptic vesicles fusing with the membrane that surrounds the cell. This “membrane fusion” process is driven by a group of proteins called the SNARE complex. Membrane fusion is triggered by a sudden increase in the amount of calcium ions in the cell, which leads to an increase in the activity of a protein called synaptotagmin-1. A region of this protein known as the C2B domain is able to detect calcium ions, and it can also bind to the cell membrane and SNARE complex proteins. However, it is not clear what roles these interactions play in driving the release of neurotransmitters. Wang, Li et al. have used a variety of biophysical techniques to study these interactions in more detail using purified proteins and other cell components. The experiments show that all three interactions occur at the same time and are all required for synaptotagmin-1 to trigger membrane fusion. Wang, Li et al. propose that these interactions allow synaptotagmin-1 to bend a section of the cell membrane in response to calcium ions. The experiments also show that the C2B domain interacts more strongly with the SNARE complex than previously thought. A future challenge is to observe whether synaptotagmin-1 works in the same way in living cells. DOI:http://dx.doi.org/10.7554/eLife.14211.002
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Gordon SE, Senning EN, Aman TK, Zagotta WN. Transition metal ion FRET to measure short-range distances at the intracellular surface of the plasma membrane. ACTA ACUST UNITED AC 2016; 147:189-200. [PMID: 26755772 PMCID: PMC4727948 DOI: 10.1085/jgp.201511530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/21/2015] [Indexed: 11/20/2022]
Abstract
A novel method is presented to measure short distances in cell plasma membranes using transition metal ion FRET with metal ions bound to introduced sites in the membrane. Biological membranes are complex assemblies of lipids and proteins that serve as platforms for cell signaling. We have developed a novel method for measuring the structure and dynamics of the membrane based on fluorescence resonance energy transfer (FRET). The method marries four technologies: (1) unroofing cells to isolate and access the cytoplasmic leaflet of the plasma membrane; (2) patch-clamp fluorometry (PCF) to measure currents and fluorescence simultaneously from a membrane patch; (3) a synthetic lipid with a metal-chelating head group to decorate the membrane with metal-binding sites; and (4) transition metal ion FRET (tmFRET) to measure short distances between a fluorescent probe and a transition metal ion on the membrane. We applied this method to measure the density and affinity of native and introduced metal-binding sites in the membrane. These experiments pave the way for measuring structural rearrangements of membrane proteins relative to the membrane.
Collapse
Affiliation(s)
- Sharona E Gordon
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Eric N Senning
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Teresa K Aman
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| |
Collapse
|
28
|
Zagotta WN, Gordon MT, Senning EN, Munari MA, Gordon SE. Measuring distances between TRPV1 and the plasma membrane using a noncanonical amino acid and transition metal ion FRET. ACTA ACUST UNITED AC 2016; 147:201-16. [PMID: 26755770 PMCID: PMC4727949 DOI: 10.1085/jgp.201511531] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/17/2015] [Indexed: 12/14/2022]
Abstract
Transition metal ion FRET between a noncanonical fluorescent amino acid incorporated into TRPV1 and metal ions bound to the cell plasma can be used to measure distances and dynamics between cytosolic domains of proteins and the membrane. Despite recent advances, the structure and dynamics of membrane proteins in cell membranes remain elusive. We implemented transition metal ion fluorescence resonance energy transfer (tmFRET) to measure distances between sites on the N-terminal ankyrin repeat domains (ARDs) of the pain-transducing ion channel TRPV1 and the intracellular surface of the plasma membrane. To preserve the native context, we used unroofed cells, and to specifically label sites in TRPV1, we incorporated a fluorescent, noncanonical amino acid, L-ANAP. A metal chelating lipid was used to decorate the plasma membrane with high-density/high-affinity metal-binding sites. The fluorescence resonance energy transfer (FRET) efficiencies between L-ANAP in TRPV1 and Co2+ bound to the plasma membrane were consistent with the arrangement of the ARDs in recent cryoelectron microscopy structures of TRPV1. No change in tmFRET was observed with the TRPV1 agonist capsaicin. These results demonstrate the power of tmFRET for measuring structure and rearrangements of membrane proteins relative to the cell membrane.
Collapse
Affiliation(s)
- William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Moshe T Gordon
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Eric N Senning
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Mika A Munari
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Sharona E Gordon
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| |
Collapse
|
29
|
Colomb W, Sarkar SK. Extracting physics of life at the molecular level: A review of single-molecule data analyses. Phys Life Rev 2015; 13:107-37. [PMID: 25660417 DOI: 10.1016/j.plrev.2015.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 12/31/2022]
Abstract
Studying individual biomolecules at the single-molecule level has proved very insightful recently. Single-molecule experiments allow us to probe both the equilibrium and nonequilibrium properties as well as make quantitative connections with ensemble experiments and equilibrium thermodynamics. However, it is important to be careful about the analysis of single-molecule data because of the noise present and the lack of theoretical framework for processes far away from equilibrium. Biomolecular motion, whether it is free in solution, on a substrate, or under force, involves thermal fluctuations in varying degrees, which makes the motion noisy. In addition, the noise from the experimental setup makes it even more complex. The details of biologically relevant interactions, conformational dynamics, and activities are hidden in the noisy single-molecule data. As such, extracting biological insights from noisy data is still an active area of research. In this review, we will focus on analyzing both fluorescence-based and force-based single-molecule experiments and gaining biological insights at the single-molecule level. Inherently nonequilibrium nature of biological processes will be highlighted. Simulated trajectories of biomolecular diffusion will be used to compare and validate various analysis techniques.
Collapse
Affiliation(s)
- Warren Colomb
- Department of Physics, Colorado School of Mines, Golden, CO 80401, United States
| | - Susanta K Sarkar
- Department of Physics, Colorado School of Mines, Golden, CO 80401, United States.
| |
Collapse
|
30
|
Azimi-Nezhad M, Lambert D, Ottone C, Perrin C, Chapel C, Gaillard G, Pfister M, Masson C, Tabone E, Betsou F, Meyronet D, Ungeheuer MN, Siest SV. Influence of pre-analytical variables on VEGF gene expression and circulating protein concentrations. Biopreserv Biobank 2015; 10:454-61. [PMID: 24845047 DOI: 10.1089/bio.2012.0016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The extended role of vascular endothelial growth factor (VEGF) in human pathophysiology led us to evaluate pre-analytical parameters possibly influencing its levels in peripheral blood and tissues. The effects on VEGF protein levels and mRNA expression were measured after storage delay (blood and tissue), use of different types of anticoagulants (blood), and after different numbers of freeze-thaw cycles (blood). METHODS Blood from healthy donors was sampled simultaneously in ethylene diamine tetraacetic acid (EDTA), acid citrate dextrose (ACD-A), hirudin, and serum separation tubes. For each anticoagulant, VEGF was measured by enzyme-linked immunosorbent assay (ELISA) with different conditions of delay at 4°C before centrifugation (2 h, 4 h, or 48 h) and of different numbers of freeze-thaw cycles (1, 2, and 10). The transcripts coding for the VEGF165 isoform were quantified in peripheral blood mononuclear cells by RT-PCR. Muscle biopsy samples were frozen with delays of 15, 30, or 60 min after surgery. VEGF expression was quantified on immunofluorescence stained slides. RESULTS The period of storage and the number of freeze-thaw cycles correlated with an increase in the levels of circulating VEGF (for each anticoagulant but not for serum) and its expression in PBMCs. VEGF expression measured from muscle biopsy sections was higher with freezing delays, with a peak at 30 and 60 min as compared to 15 min. CONCLUSIONS The most reliable conditions for measuring both circulating VEGF and its gene expression are to reduce time between blood collection and centrifugation, and to avoid multiple freeze-thaw cycles. Serum collection tubes with no additive and no separator were less sensitive to the pre-analytical variations analyzed in this study. Freezing delay had a significant influence on VEGF protein expression in tissue samples.
Collapse
Affiliation(s)
- Mohsen Azimi-Nezhad
- 1 Unité de Recherche "Génétique Cardiovasculaire", Université de Lorraine , Nancy, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kusch J, Zifarelli G. Patch-clamp fluorometry: electrophysiology meets fluorescence. Biophys J 2014; 106:1250-7. [PMID: 24655500 DOI: 10.1016/j.bpj.2014.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/23/2013] [Accepted: 02/06/2014] [Indexed: 12/26/2022] Open
Abstract
Ion channels and transporters are membrane proteins whose functions are driven by conformational changes. Classical biophysical techniques provide insight into either the structure or the function of these proteins, but a full understanding of their behavior requires a correlation of both these aspects in time. Patch-clamp and voltage-clamp fluorometry combine spectroscopic and electrophysiological techniques to simultaneously detect conformational changes and ionic currents across the membrane. Since its introduction, patch-clamp fluorometry has been responsible for invaluable advances in our knowledge of ion channel biophysics. Over the years, the technique has been applied to many different ion channel families to address several biophysical questions with a variety of spectroscopic approaches and electrophysiological configurations. This review illustrates the strength and the flexibility of patch-clamp fluorometry, demonstrating its potential as a tool for future research.
Collapse
Affiliation(s)
- Jana Kusch
- Universitätsklinikum Jena, Institut für Physiologie II, Jena, Germany.
| | - Giovanni Zifarelli
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genova, Italy.
| |
Collapse
|
32
|
Connolly NMC, Prehn JHM. The metabolic response to excitotoxicity - lessons from single-cell imaging. J Bioenerg Biomembr 2014; 47:75-88. [PMID: 25262286 DOI: 10.1007/s10863-014-9578-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/26/2014] [Indexed: 12/26/2022]
Abstract
Excitotoxicity is a pathological process implicated in neuronal death during ischaemia, traumatic brain injuries and neurodegenerative diseases. Excitotoxicity is caused by excess levels of glutamate and over-activation of NMDA or calcium-permeable AMPA receptors on neuronal membranes, leading to ionic influx, energetic stress and potential neuronal death. The metabolic response of neurons to excitotoxicity is complex and plays a key role in the ability of the neuron to adapt and recover from such an insult. Single-cell imaging is a powerful experimental technique that can be used to study the neuronal metabolic response to excitotoxicity in vitro and, increasingly, in vivo. Here, we review some of the knowledge of the neuronal metabolic response to excitotoxicity gained from in vitro single-cell imaging, including calcium and ATP dynamics and their effects on mitochondrial function, along with the contribution of glucose metabolism, oxidative stress and additional neuroprotective signalling mechanisms. Future work will combine knowledge gained from single-cell imaging with data from biochemical and computational techniques to garner holistic information about the metabolic response to excitotoxicity at the whole brain level and transfer this knowledge to a clinical setting.
Collapse
Affiliation(s)
- Niamh M C Connolly
- Department of Physiology and Medical Physics, 123 St Stephen's Green, Dublin 2, Ireland
| | | |
Collapse
|
33
|
Bosmans F. New rule(r)s for FRET. Biophys J 2014; 105:2619-20. [PMID: 24359733 DOI: 10.1016/j.bpj.2013.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 11/05/2013] [Indexed: 11/15/2022] Open
Affiliation(s)
- Frank Bosmans
- Department of Physiology and Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
34
|
Kang HS, Kim J, Lee HJ, Kwon BM, Lee DK, Hong SH. LRP1-dependent pepsin clearance induced by 2'-hydroxycinnamaldehyde attenuates breast cancer cell invasion. Int J Biochem Cell Biol 2014; 53:15-23. [PMID: 24796846 DOI: 10.1016/j.biocel.2014.04.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/08/2014] [Accepted: 04/26/2014] [Indexed: 12/20/2022]
Abstract
2'-Hydroxycinnamaldehyde inhibits breast cancer cell invasion. This study examined whether 2'-hydroxycinnamaldehyde, acting as a Michael acceptor, interferes with the ligand binding of low-density lipoprotein receptor-related protein 1 to mediate breast cancer cell invasion. Low-density lipoprotein receptor-related protein 1, one of the direct molecular targets of 2'-hydroxycinnamaldehyde, is a multifunctional endocytic receptor. Changes in the thiol oxidation status of cell surface receptor proteins may function as a molecular switch, influencing ligand(s) binding. The oxidation status of extracellular cysteine thiol groups in MCF-7 and MDA-MB-231 cells was examined using a fluorescence-activated cell sorter with thiol-specific fluorescent probes; Matrigel invasion and wound-healing assays were performed to determine the effects of 2'-hydroxycinnamaldehyde on in vitro cell migration. The molecular mechanisms by which 2'-hydroxycinnamaldehyde acts were evaluated by transient knockdown using siRNA or inhibition of low-density lipoprotein receptor-related protein 1 by receptor-associated protein treatment. 2'-Hydroxycinnamaldehyde increased α-2-macroglobulin binding to low-density lipoprotein receptor-related protein 1, which was alleviated by pretreatment of cells with N-acetylcystein. 2'-Hydroxycinnamaldehyde decreased the extracellular pepsin concentration significantly in a low-density lipoprotein receptor-related protein 1- and α-2-macroglobulin-dependent manner. The anti-invasive effect of 2'-hydroxycinnamaldehyde was mitigated with receptor-associated protein pretreatment, suggesting that low-density lipoprotein receptor-related protein 1 is essential for the effects of 2'-hydroxycinnamaldehyde. From these data, we suggest that 2'-hydroxycinnamaldehyde increases the cysteine thiol oxidation status of low-density lipoprotein receptor-related protein 1 extracellular domains, which results in α-2-macroglobulin ligand binding stimulation. Therefore, pepsin clearance in a low-density lipoprotein receptor-related protein 1-α-2-macroglobulin-dependent manner might be an important molecular mechanism in 2'-hydroxycinnamaldehyde exerting its anti-invasive action on breast cancer cells. Furthermore, our data may provide an opportunity to promote the importance of the thiol oxidation status of cell surface receptor proteins for regulating cellular signaling pathways that are important in cancer progression.
Collapse
Affiliation(s)
- Hye Suk Kang
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea; Global Research Laboratory for RNAi Medicine, Department of Chemistry, Sungkyunkwan University, Suwon 440-746, South Korea
| | - JinKyoung Kim
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea
| | - Heon-Jin Lee
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, University of Science and Technology in Korea, Daejon 305-806, South Korea
| | - Dong-Ki Lee
- Global Research Laboratory for RNAi Medicine, Department of Chemistry, Sungkyunkwan University, Suwon 440-746, South Korea
| | - Su-Hyung Hong
- Department of Oral Microbiology, School of Dentistry, Kyungpook National University, Daegu 700-412, South Korea.
| |
Collapse
|
35
|
HTS assays for developing the molecular pharmacology of ion channels. Curr Opin Pharmacol 2014; 15:91-6. [DOI: 10.1016/j.coph.2014.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 01/29/2023]
|
36
|
Crystal structure of the plant dual-affinity nitrate transporter NRT1.1. Nature 2014; 507:73-7. [PMID: 24572362 PMCID: PMC3968801 DOI: 10.1038/nature13074] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 01/23/2014] [Indexed: 01/24/2023]
Abstract
Nitrate is a primary nutrient for plant growth, but its levels in soil can fluctuate by several orders of magnitude. Previous studies have identified Arabidopsis NRT1.1 as a dual-affinity nitrate transporter, which can take up nitrate over a wide range of concentrations. The mode of action of NRT1.1 is controlled by phosphorylation of a key residue, Thr101. Yet how this posttranslational modification switches the transporter between two affinity states remains unclear. Here we report the crystal structure of unphosphorylated NRT1.1, which reveals an unexpected homodimer in the inward-facing conformation. In this low-affinity state, the Thr101 phosphorylation site is embedded in a pocket immediately adjacent to the dimer interface, linking the phosphorylation status of the transporter to its oligomeric state. Using a cell-based fluorescence resonance energy transfer assay, we show that functional NRT1.1 indeed dimerizes in the cell membrane and the phosphomimetic mutation of Thr101 converts the protein into a monophasic high affinity transporter by structurally decoupling the dimer. Together with analyses of the substrate transport tunnel, our results establish a phosphorylation-controlled dimerization switch that allows NRT1.1 to uptake nitrate with two distinct affinity modes.
Collapse
|
37
|
Intraoperative fluorescent imaging of intracranial tumors: a review. Clin Neurol Neurosurg 2013; 115:517-28. [PMID: 23523009 DOI: 10.1016/j.clineuro.2013.02.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/15/2013] [Accepted: 02/18/2013] [Indexed: 11/24/2022]
Abstract
A review of fluorescent imaging for intracranial neoplasms is presented. Complete resection of brain cancer is seldom possible because of the goal to preserve brain tissue and the inability to visualize individual infiltrative tumor cells. Verification of histology and identification of tumor invasion in macroscopically normal-appearing brain tissue determine prognosis after resection of malignant gliomas. Therefore, imaging modalities aim to facilitate intraoperative decision-making. Intraoperative fluorescent imaging techniques have the potential to enable precise histopathologic diagnosis and to detect tumor remnants in the operative field. Macroscopic fluorescence imaging is effective for gross tumor detection. Microscopic imaging techniques enhance the sensitivity of the macroscopic observations and provide real-time histological information. Further development of clinical grade fluorescent agents specifically targeting tumor cells could improve the diagnostic and prognostic yield of intraoperative imaging.
Collapse
|
38
|
Yu X, Wu X, Bermejo GA, Brooks BR, Taraska JW. Accurate high-throughput structure mapping and prediction with transition metal ion FRET. Structure 2012; 21:9-19. [PMID: 23273426 DOI: 10.1016/j.str.2012.11.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/15/2012] [Accepted: 11/17/2012] [Indexed: 11/28/2022]
Abstract
Mapping the landscape of a protein's conformational space is essential to understanding its functions and regulation. The limitations of many structural methods have made this process challenging for most proteins. Here, we report that transition metal ion FRET (tmFRET) can be used in a rapid, highly parallel screen, to determine distances from multiple locations within a protein at extremely low concentrations. The distances generated through this screen for the protein maltose binding protein (MBP) match distances from the crystal structure to within a few angstroms. Furthermore, energy transfer accurately detects structural changes during ligand binding. Finally, fluorescence-derived distances can be used to guide molecular simulations to find low energy states. Our results open the door to rapid, accurate mapping and prediction of protein structures at low concentrations, in large complex systems, and in living cells.
Collapse
Affiliation(s)
- Xiaozhen Yu
- Laboratory of Molecular Biophysics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiongwu Wu
- Laboratory of Computational Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guillermo A Bermejo
- Division of Computational Bioscience, Center for Information Technology, National Institutes of Health, Bethesda, MD 20892, USA
| | - Bernard R Brooks
- Laboratory of Computational Biology, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
39
|
Puljung MC, Zagotta WN. Fluorescent labeling of specific cysteine residues using CyMPL. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2012; Chapter 14:14.14.1-14.14.10. [PMID: 23151742 PMCID: PMC3501994 DOI: 10.1002/0471140864.ps1414s70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The unique reactivity and relative rarity of cysteine among amino acids makes it a convenient target for the site-specific chemical modification of proteins. Commercially available fluorophores and modifiers react with cysteine through a variety of electrophilic functional groups. However, it can be difficult to achieve specific labeling of a particular cysteine residue in a protein containing multiple cysteines, in a mixture of proteins, or in a protein's native environment. This unit describes a procedure termed CyMPL (Cysteine Metal Protection and Labeling), which enables specific labeling by incorporating a cysteine of interest into a minimal binding site for group 12 metal ions (e.g., Cd2+ and Zn2+). These sites can be inserted into any region of known secondary structure in virtually any protein and cause minimal structural perturbation. Bound metal ions protect the cysteine from reaction while background cysteines are covalently blocked with non-fluorescent modifiers. The metal ions are subsequently removed and the deprotected cysteine is labeled specifically.
Collapse
Affiliation(s)
- Michael C Puljung
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| | - William N Zagotta
- Department of Physiology and Biophysics, University of Washington, Seattle, Washington
| |
Collapse
|
40
|
Blunck R, Batulan Z. Mechanism of electromechanical coupling in voltage-gated potassium channels. Front Pharmacol 2012; 3:166. [PMID: 22988442 PMCID: PMC3439648 DOI: 10.3389/fphar.2012.00166] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/24/2012] [Indexed: 01/10/2023] Open
Abstract
Voltage-gated ion channels play a central role in the generation of action potentials in the nervous system. They are selective for one type of ion - sodium, calcium, or potassium. Voltage-gated ion channels are composed of a central pore that allows ions to pass through the membrane and four peripheral voltage sensing domains that respond to changes in the membrane potential. Upon depolarization, voltage sensors in voltage-gated potassium channels (Kv) undergo conformational changes driven by positive charges in the S4 segment and aided by pairwise electrostatic interactions with the surrounding voltage sensor. Structure-function relations of Kv channels have been investigated in detail, and the resulting models on the movement of the voltage sensors now converge to a consensus; the S4 segment undergoes a combined movement of rotation, tilt, and vertical displacement in order to bring 3-4e(+) each through the electric field focused in this region. Nevertheless, the mechanism by which the voltage sensor movement leads to pore opening, the electromechanical coupling, is still not fully understood. Thus, recently, electromechanical coupling in different Kv channels has been investigated with a multitude of techniques including electrophysiology, 3D crystal structures, fluorescence spectroscopy, and molecular dynamics simulations. Evidently, the S4-S5 linker, the covalent link between the voltage sensor and pore, plays a crucial role. The linker transfers the energy from the voltage sensor movement to the pore domain via an interaction with the S6 C-termini, which are pulled open during gating. In addition, other contact regions have been proposed. This review aims to provide (i) an in-depth comparison of the molecular mechanisms of electromechanical coupling in different Kv channels; (ii) insight as to how the voltage sensor and pore domain influence one another; and (iii) theoretical predictions on the movement of the cytosolic face of the Kv channels during gating.
Collapse
Affiliation(s)
- Rikard Blunck
- Groupe d’étude des protéines membranairesMontreal, QC, Canada
- Department of Physiology, Université de MontréalMontreal, QC, Canada
- Department of Physics, Université de MontréalMontreal, QC, Canada
| | - Zarah Batulan
- Groupe d’étude des protéines membranairesMontreal, QC, Canada
- Department of Physiology, Université de MontréalMontreal, QC, Canada
| |
Collapse
|
41
|
Iourov IY, Vorsanova SG, Yurov YB. Single cell genomics of the brain: focus on neuronal diversity and neuropsychiatric diseases. Curr Genomics 2012; 13:477-88. [PMID: 23449087 PMCID: PMC3426782 DOI: 10.2174/138920212802510439] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 01/30/2012] [Accepted: 06/12/2012] [Indexed: 12/21/2022] Open
Abstract
Single cell genomics has made increasingly significant contributions to our understanding of the role that somatic genome variations play in human neuronal diversity and brain diseases. Studying intercellular genome and epigenome variations has provided new clues to the delineation of molecular mechanisms that regulate development, function and plasticity of the human central nervous system (CNS). It has been shown that changes of genomic content and epigenetic profiling at single cell level are involved in the pathogenesis of neuropsychiatric diseases (schizophrenia, mental retardation (intellectual/leaning disability), autism, Alzheimer's disease etc.). Additionally, several brain diseases were found to be associated with genome and chromosome instability (copy number variations, aneuploidy) variably affecting cell populations of the human CNS. The present review focuses on the latest advances of single cell genomics, which have led to a better understanding of molecular mechanisms of neuronal diversity and neuropsychiatric diseases, in the light of dynamically developing fields of systems biology and "omics".
Collapse
Affiliation(s)
- Ivan Y Iourov
- National Research Center of Mental Health, Russian Academy of Medical Sciences, Moscow, Russia
- Institute of Pediatrics and Children Surgery, Minzdravsotsrazvitia, Moscow, Russia
| | - Svetlana G Vorsanova
- National Research Center of Mental Health, Russian Academy of Medical Sciences, Moscow, Russia
- Institute of Pediatrics and Children Surgery, Minzdravsotsrazvitia, Moscow, Russia
- Center for Neurobiological Diagnosis of Genetic Psychiatric Disorders, Moscow City University of Psychology and Education, Russia
| | - Yuri B Yurov
- National Research Center of Mental Health, Russian Academy of Medical Sciences, Moscow, Russia
- Institute of Pediatrics and Children Surgery, Minzdravsotsrazvitia, Moscow, Russia
- Center for Neurobiological Diagnosis of Genetic Psychiatric Disorders, Moscow City University of Psychology and Education, Russia
| |
Collapse
|
42
|
Richards CI, Luong K, Srinivasan R, Turner SW, Dougherty DA, Korlach J, Lester HA. Live-cell imaging of single receptor composition using zero-mode waveguide nanostructures. NANO LETTERS 2012; 12:3690-4. [PMID: 22668081 PMCID: PMC3397148 DOI: 10.1021/nl301480h] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
We exploit the optical and spatial features of subwavelength nanostructures to examine individual receptors on the plasma membrane of living cells. Receptors were sequestered in portions of the membrane projected into zero-mode waveguides. Using single-step photobleaching of green fluorescent protein incorporated into individual subunits, the resulting spatial isolation was used to measure subunit stoichiometry in α4β4 and α4β2 nicotinic acetylcholine and P2X2 ATP receptors. We also show that nicotine and cytisine have differential effects on α4β2 stoichiometry.
Collapse
Affiliation(s)
- Christopher I. Richards
- Division of Biology 156-29, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125
- Department of Chemistry, University of Kentucky, Chemistry-Physics Building, Lexington, KY 40506
| | - Khai Luong
- Pacific Biosciences, 1380 Willow Road, Menlo Park, CA 94025
| | - Rahul Srinivasan
- Division of Biology 156-29, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125
| | | | - Dennis A. Dougherty
- Division of Chemistry & Chemical Engineering 164-30, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125
| | - Jonas Korlach
- Pacific Biosciences, 1380 Willow Road, Menlo Park, CA 94025
| | - Henry A. Lester
- Division of Biology 156-29, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91125
| |
Collapse
|
43
|
Long XB, Hu S, Wang N, Zhen HT, Cui YH, Liu Z. Clara cell 10-kDa protein gene transfection inhibits NF-κB activity in airway epithelial cells. PLoS One 2012; 7:e35960. [PMID: 22558282 PMCID: PMC3338482 DOI: 10.1371/journal.pone.0035960] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 03/24/2012] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Clara cell 10-kDa protein (CC10) is a multifunctional protein with anti-inflammatory and immunomodulatory effects. Induction of CC10 expression by gene transfection may possess potential therapeutic effect. Nuclear factor κB (NF-κB) plays a key role in the inflammatory processes of airway diseases. METHOD/RESULTS To investigate potential therapeutic effect of CC10 gene transfection in controlling airway inflammation and the underlying intracellular mechanisms, in this study, we constructed CC10 plasmid and transfected it into bronchial epithelial cell line BEAS-2B cells and CC10 knockout mice. In BEAS-2B cells, CC10's effect on interleukin (IL)-1β induced IL-8 expression was explored by means of RT-PCR and ELISA and its effect on NF-κB classical signaling pathway was studied by luciferase reporter, western blot, and immunoprecipitation assay. The effect of endogenous CC10 on IL-1β evoked IL-8 expression was studied by means of nasal explant culture. In mice, CC10's effect on IL-1β induced IL-8 and nuclear p65 expression was examined by immunohistochemistry. First, we found that the CC10 gene transfer could inhibit IL-1β induced IL-8 expression in BEAS-2B cells. Furthermore, we found that CC10 repressed IL-1β induced NF-κB activation by inhibiting the phosphorylation of IκB-α but not IκB kinase-α/β in BEAS-2B cells. Nevertheless, we did not observe a direct interaction between CC10 and p65 subunit in BEAS-2B cells. In nasal explant culture, we found that IL-1β induced IL-8 expression was inversely correlated with CC10 levels in human sinonasal mucosa. In vivo study revealed that CC10 gene transfer could attenuate the increase of IL-8 and nuclear p65 staining in nasal epithelial cells in CC10 knockout mice evoked by IL-1β administration. CONCLUSION These results indicate that CC10 gene transfer may inhibit airway inflammation through suppressing the activation of NF-κB, which may provide us a new consideration in the therapy of airway inflammation.
Collapse
Affiliation(s)
- Xiao-Bo Long
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Shuang Hu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Nan Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hong-Tao Zhen
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yong-Hua Cui
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
44
|
Ganguli S, Sompolinsky H. Compressed sensing, sparsity, and dimensionality in neuronal information processing and data analysis. Annu Rev Neurosci 2012; 35:485-508. [PMID: 22483042 DOI: 10.1146/annurev-neuro-062111-150410] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The curse of dimensionality poses severe challenges to both technical and conceptual progress in neuroscience. In particular, it plagues our ability to acquire, process, and model high-dimensional data sets. Moreover, neural systems must cope with the challenge of processing data in high dimensions to learn and operate successfully within a complex world. We review recent mathematical advances that provide ways to combat dimensionality in specific situations. These advances shed light on two dual questions in neuroscience. First, how can we as neuroscientists rapidly acquire high-dimensional data from the brain and subsequently extract meaningful models from limited amounts of these data? And second, how do brains themselves process information in their intrinsically high-dimensional patterns of neural activity as well as learn meaningful, generalizable models of the external world from limited experience?
Collapse
Affiliation(s)
- Surya Ganguli
- Department of Applied Physics, Stanford University, Stanford, California 94305, USA.
| | | |
Collapse
|
45
|
Taraska JW. Mapping membrane protein structure with fluorescence. Curr Opin Struct Biol 2012; 22:507-13. [PMID: 22445227 DOI: 10.1016/j.sbi.2012.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 02/22/2012] [Accepted: 02/24/2012] [Indexed: 01/07/2023]
Abstract
Membrane proteins regulate many cellular processes including signaling cascades, ion transport, membrane fusion, and cell-to-cell communications. Understanding the architecture and conformational fluctuations of these proteins is critical to understanding their regulation and functions. Fluorescence methods including intensity mapping, fluorescence resonance energy transfer (FRET), and photo-induced electron transfer, allow for targeted measurements of domains within membrane proteins. These methods can reveal how a protein is structured and how it transitions between different conformational states. Here, I will review recent work done using fluorescence to map the structures of membrane proteins, focusing on how each of these methods can be applied to understanding the dynamic nature of individual membrane proteins and protein complexes.
Collapse
Affiliation(s)
- Justin W Taraska
- Laboratory of Molecular Biophysics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
46
|
Lamy CM, Sallin O, Loussert C, Chatton JY. Sodium sensing in neurons with a dendrimer-based nanoprobe. ACS NANO 2012; 6:1176-1187. [PMID: 22288942 DOI: 10.1021/nn203822t] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Ion imaging is a powerful methodology to assess fundamental biological processes in live cells. The limited efficiency of some ion-sensing probes and their fast leakage from cells are important restrictions to this approach. In this study, we present a novel strategy based on the use of dendrimer nanoparticles to obtain better intracellular retention of fluorescent probes and perform prolonged fluorescence imaging of intracellular ion dynamics. A new sodium-sensitive nanoprobe was generated by encapsulating a sodium dye in a PAMAM dendrimer nanocontainer. This nanoprobe is very stable and has high sodium sensitivity and selectivity. When loaded in neurons in live brain tissue, it homogenously fills the entire cell volume, including small processes, and stays for long durations, with no detectable alterations of cell functional properties. We demonstrate the suitability of this new sodium nanosensor for monitoring physiological sodium responses such as those occurring during neuronal activity.
Collapse
Affiliation(s)
- Christophe M Lamy
- Department of Cell Biology and Morphology, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland.
| | | | | | | |
Collapse
|
47
|
Gauthier-Kemper A, Weissmann C, Reyher HJ, Brandt R. Monitoring Cytoskeletal Dynamics in Living Neurons Using Fluorescence Photoactivation. Methods Enzymol 2012; 505:3-21. [DOI: 10.1016/b978-0-12-388448-0.00009-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
48
|
Jakobsson J, Ackermann F, Andersson F, Larhammar D, Löw P, Brodin L. Regulation of synaptic vesicle budding and dynamin function by an EHD ATPase. J Neurosci 2011; 31:13972-80. [PMID: 21957258 PMCID: PMC6633164 DOI: 10.1523/jneurosci.1289-11.2011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 08/01/2011] [Accepted: 08/09/2011] [Indexed: 12/24/2022] Open
Abstract
Eps15 homology domain-containing proteins (EHDs) are conserved ATPases implicated in membrane remodeling. Recently, EHD1 was found to be enriched at synaptic release sites, suggesting a possible involvement in the trafficking of synaptic vesicles. We have investigated the role of an EHD1/3 ortholog (l-EHD) in the lamprey giant reticulospinal synapse. l-EHD was detected by immunogold at endocytic structures adjacent to release sites. In antibody microinjection experiments, perturbation of l-EHD inhibited synaptic vesicle endocytosis and caused accumulation of clathrin-coated pits with atypical, elongated necks. The necks were covered with helix-like material containing dynamin. To test whether l-EHD directly interferes with dynamin function, we used fluid-supported bilayers as in vitro assay. We found that l-EHD strongly inhibited vesicle budding induced by dynamin in the constant presence of GTP. l-EHD also inhibited dynamin-induced membrane tubulation in the presence of GTPγS, a phenomenon linked with dynamin helix assembly. Our in vivo results demonstrate the involvement of l-EHD in clathrin/dynamin-dependent synaptic vesicle budding. Based on our in vitro observations, we suggest that l-EHD acts to limit the formation of long, unproductive dynamin helices, thereby promoting vesicle budding.
Collapse
Affiliation(s)
- Joel Jakobsson
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden, and
| | - Frauke Ackermann
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden, and
| | - Fredrik Andersson
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden, and
| | - Dan Larhammar
- Department of Neuroscience, Uppsala University, S-751 24, Uppsala, Sweden
| | - Peter Löw
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden, and
| | - Lennart Brodin
- Department of Neuroscience, Karolinska Institutet, S-171 77, Stockholm, Sweden, and
| |
Collapse
|
49
|
Crivat G, Taraska JW. Imaging proteins inside cells with fluorescent tags. Trends Biotechnol 2011; 30:8-16. [PMID: 21924508 DOI: 10.1016/j.tibtech.2011.08.002] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 08/04/2011] [Accepted: 08/04/2011] [Indexed: 11/30/2022]
Abstract
Watching biological molecules provides clues to their function and regulation. Some of the most powerful methods of labeling proteins for imaging use genetically encoded fluorescent fusion tags. There are four standard genetic methods of covalently tagging a protein with a fluorescent probe for cellular imaging. These use (i) autofluorescent proteins, (ii) self-labeling enzymes, (iii) enzymes that catalyze the attachment of a probe to a target sequence, and (iv) biarsenical dyes that target tetracysteine motifs. Each of these techniques has advantages and disadvantages. In this review, we cover new developments in these methods and discuss practical considerations for their use in imaging proteins inside living cells.
Collapse
Affiliation(s)
- Georgeta Crivat
- University of Maryland, College Park, Department of Entomology and Program in Cell and Molecular Biology, College Park, MD 20742, USA
| | | |
Collapse
|
50
|
Labeling of specific cysteines in proteins using reversible metal protection. Biophys J 2011; 100:2513-21. [PMID: 21575586 DOI: 10.1016/j.bpj.2011.03.063] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 02/14/2011] [Accepted: 03/29/2011] [Indexed: 02/05/2023] Open
Abstract
Fluorescence spectroscopy is an indispensible tool for studying the structure and conformational dynamics of protein molecules both in isolation and in their cellular context. The ideal probes for monitoring intramolecular protein motions are small, cysteine-reactive fluorophores. However, it can be difficult to obtain specific labeling of a desired cysteine in proteins with multiple cysteines, in a mixture of proteins, or in a protein's native environment, in which many cysteine-containing proteins are present. To obtain specific labeling, we developed a method we call cysteine metal protection and labeling (CyMPL). With this method, a desired cysteine can be reversibly protected by binding group 12 metal ions (e.g., Cd²⁺ and Zn²⁺) while background cysteines are blocked with nonfluorescent covalent modifiers. We increased the metal affinity for specific cysteines by incorporating them into minimal binding sites in existing secondary structural motifs (i.e., α-helix or β-strand). After the metal ions were removed, the deprotected cysteines were then available to specifically react with a fluorophore.
Collapse
|