1
|
Lutomski CA, Bennett JL, El-Baba TJ, Wu D, Hinkle JD, Burnap SA, Liko I, Mullen C, Syka JEP, Struwe WB, Robinson CV. Defining proteoform-specific interactions for drug targeting in a native cell signalling environment. Nat Chem 2025; 17:204-214. [PMID: 39806141 PMCID: PMC11794133 DOI: 10.1038/s41557-024-01711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025]
Abstract
Understanding the dynamics of membrane protein-ligand interactions within a native lipid bilayer is a major goal for drug discovery. Typically, cell-based assays are used, however, they are often blind to the effects of protein modifications. In this study, using the archetypal G protein-coupled receptor rhodopsin, we found that the receptor and its effectors can be released directly from retina rod disc membranes using infrared irradiation in a mass spectrometer. Subsequent isolation and dissociation by infrared multiphoton dissociation enabled the sequencing of individual retina proteoforms. Specifically, we categorized distinct proteoforms of rhodopsin, localized labile palmitoylations, discovered a Gβγ proteoform that abolishes membrane association and defined lipid modifications on G proteins that influence their assembly. Given reports of undesirable side-effects involving vision, we characterized the off-target drug binding of two phosphodiesterase 5 inhibitors, vardenafil and sildenafil, to the retina rod phosphodiesterase 6 (PDE6). The results demonstrate differential off-target reactivity with PDE6 and an interaction preference for lipidated proteoforms of G proteins. In summary, this study highlights the opportunities for probing proteoform-ligand interactions within natural membrane environments.
Collapse
Affiliation(s)
- Corinne A Lutomski
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Jack L Bennett
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Tarick J El-Baba
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Di Wu
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | | | - Sean A Burnap
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | | | | | | | - Weston B Struwe
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Carol V Robinson
- Physical and Theoretical Chemistry Laboratory, Department of Chemistry, University of Oxford, Oxford, UK.
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Ashok S, Ramachandra Rao S. Updates on protein-prenylation and associated inherited retinopathies. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1410874. [PMID: 39026984 PMCID: PMC11254824 DOI: 10.3389/fopht.2024.1410874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Membrane-anchored proteins play critical roles in cell signaling, cellular architecture, and membrane biology. Hydrophilic proteins are post-translationally modified by a diverse range of lipid molecules such as phospholipids, glycosylphosphatidylinositol, and isoprenes, which allows their partition and anchorage to the cell membrane. In this review article, we discuss the biochemical basis of isoprenoid synthesis, the mechanisms of isoprene conjugation to proteins, and the functions of prenylated proteins in the neural retina. Recent discovery of novel prenyltransferases, prenylated protein chaperones, non-canonical prenylation-target motifs, and reversible prenylation is expected to increase the number of inherited systemic and blinding diseases with aberrant protein prenylation. Recent important investigations have also demonstrated the role of several unexpected regulators (such as protein charge, sequence/protein-chaperone interaction, light exposure history) in the photoreceptor trafficking of prenylated proteins. Technical advances in the investigation of the prenylated proteome and its application in vision research are discussed. Clinical updates and technical insights into known and putative prenylation-associated retinopathies are provided herein. Characterization of non-canonical prenylation mechanisms in the retina and retina-specific prenylated proteome is fundamental to the understanding of the pathogenesis of protein prenylation-associated inherited blinding disorders.
Collapse
Affiliation(s)
- Sudhat Ashok
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
| | - Sriganesh Ramachandra Rao
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, State University of New York, University at Buffalo, Buffalo, NY, United States
- Research Service, VA Western New York Healthcare System, Buffalo, NY, United States
| |
Collapse
|
3
|
Sun W, Chen Y, Yang Y, Wang P, Gong J, Han X, Xu C, Luan H, Li S, Li R, Wen B, Lv S, Wei C. Characteristics and Transcriptomic Analysis of Cholinergic Neurons Derived from Induced Pluripotent Stem Cells with APP Mutation in Alzheimer's Disease. J Alzheimers Dis 2024; 101:637-649. [PMID: 39213067 DOI: 10.3233/jad-240299] [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: 09/04/2024]
Abstract
Background The cholinergic hypothesis is one of the main theories that describe the pathogenesis of Alzheimer's disease (AD). Cholinergic neurons degenerate early and are severely damaged in AD. Despite extensive research, the causes of cholinergic neuron damage and the underlying molecular changes remain unclear. Objective This study aimed to explore the characteristics and transcriptomic changes in cholinergic neurons derived from human induced pluripotent stem cells (iPSCs) with APP mutation. Methods Peripheral blood mononuclear cells from patients with AD and healthy individuals were reprogrammed into iPSCs. The iPSCs were differentiated into cholinergic neurons. Cholinergic neurons were stained, neurotoxically tested, and electrophysiologically and transcriptomically analyzed. Results The iPSCs-derived cholinergic neurons from a patient with AD carrying a mutation in APP displayed enhanced susceptibility to Aβ1-42-induced neurotoxicity, characterized by severe neurotoxic effects, such as cell body coagulation and neurite fragmentation. Cholinergic neurons exhibited electrophysiological impairments and neuronal death after 21 days of culture in the AD group. Transcriptome analysis disclosed 883 differentially expressed genes (DEGs, 420 upregulated and 463 downregulated) participating in several signaling pathways implicated in AD pathogenesis. To assess the reliability of RNA sequencing, the expression of 16 target DEGs was validated using qPCR. Finally, the expression of the 8 core genes in different cell types of brain was analyzed by the AlzData database. Conclusions In this study, iPSCs-derived cholinergic neurons from AD patients with APP mutations exhibit characteristics reminiscent of neurodegenerative disease. Transcriptome analysis revealed the corresponding DEGs and pathways, providing potential biomarkers and therapeutic targets for advancing AD research.
Collapse
Affiliation(s)
- Wenxian Sun
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yufei Chen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yuting Yang
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Pin Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jin Gong
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaodong Han
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Chang Xu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Heya Luan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Shaoqi Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ruina Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Boye Wen
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Sirong Lv
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Cuibai Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
4
|
Kolesnikov AV, Lobysheva E, Gnana-Prakasam JP, Kefalov VJ, Kisselev OG. Regulation of rod photoreceptor function by farnesylated G-protein γ-subunits. PLoS One 2022; 17:e0272506. [PMID: 35939447 PMCID: PMC9359561 DOI: 10.1371/journal.pone.0272506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022] Open
Abstract
Heterotrimeric G-protein transducin, Gt, is a key signal transducer and amplifier in retinal rod and cone photoreceptor cells. Despite similar subunit composition, close amino acid identity, and identical posttranslational farnesylation of their Gγ subunits, rods and cones rely on unique Gγ1 (Gngt1) and Gγc (Gngt2) isoforms, respectively. The only other farnesylated G-protein γ-subunit, Gγ11 (Gng11), is expressed in multiple tissues but not retina. To determine whether Gγ1 regulates uniquely rod phototransduction, we generated transgenic rods expressing Gγ1, Gγc, or Gγ11 in Gγ1-deficient mice and analyzed their properties. Immunohistochemistry and Western blotting demonstrated the robust expression of each transgenic Gγ in rod cells and restoration of Gαt1 expression, which is greatly reduced in Gγ1-deficient rods. Electroretinography showed restoration of visual function in all three transgenic Gγ1-deficient lines. Recordings from individual transgenic rods showed that photosensitivity impaired in Gγ1-deficient rods was also fully restored. In all dark-adapted transgenic lines, Gαt1 was targeted to the outer segments, reversing its diffuse localization found in Gγ1-deficient rods. Bright illumination triggered Gαt1 translocation from the rod outer to inner segments in all three transgenic strains. However, Gαt1 translocation in Gγ11 transgenic mice occurred at significantly dimmer background light. Consistent with this, transretinal ERG recordings revealed gradual response recovery in moderate background illumination in Gγ11 transgenic mice but not in Gγ1 controls. Thus, while farnesylated Gγ subunits are functionally active and largely interchangeable in supporting rod phototransduction, replacement of retina-specific Gγ isoforms by the ubiquitous Gγ11 affects the ability of rods to adapt to background light.
Collapse
Affiliation(s)
- Alexander V. Kolesnikov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Elena Lobysheva
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jaya P. Gnana-Prakasam
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Vladimir J. Kefalov
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, CA, United States of America
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Physiology and Biophysics, University of California, Irvine, CA, United States of America
| | - Oleg G. Kisselev
- Department of Ophthalmology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| |
Collapse
|
5
|
Functional compartmentalization of photoreceptor neurons. Pflugers Arch 2021; 473:1493-1516. [PMID: 33880652 DOI: 10.1007/s00424-021-02558-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Retinal photoreceptors are neurons that convert dynamically changing patterns of light into electrical signals that are processed by retinal interneurons and ultimately transmitted to vision centers in the brain. They represent the essential first step in seeing without which the remainder of the visual system is rendered moot. To support this role, the major functions of photoreceptors are segregated into three main specialized compartments-the outer segment, the inner segment, and the pre-synaptic terminal. This compartmentalization is crucial for photoreceptor function-disruption leads to devastating blinding diseases for which therapies remain elusive. In this review, we examine the current understanding of the molecular and physical mechanisms underlying photoreceptor functional compartmentalization and highlight areas where significant knowledge gaps remain.
Collapse
|
6
|
Barnes CL, Malhotra H, Calvert PD. Compartmentalization of Photoreceptor Sensory Cilia. Front Cell Dev Biol 2021; 9:636737. [PMID: 33614665 PMCID: PMC7889997 DOI: 10.3389/fcell.2021.636737] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Functional compartmentalization of cells is a universal strategy for segregating processes that require specific components, undergo regulation by modulating concentrations of those components, or that would be detrimental to other processes. Primary cilia are hair-like organelles that project from the apical plasma membranes of epithelial cells where they serve as exclusive compartments for sensing physical and chemical signals in the environment. As such, molecules involved in signal transduction are enriched within cilia and regulating their ciliary concentrations allows adaptation to the environmental stimuli. The highly efficient organization of primary cilia has been co-opted by major sensory neurons, olfactory cells and the photoreceptor neurons that underlie vision. The mechanisms underlying compartmentalization of cilia are an area of intense current research. Recent findings have revealed similarities and differences in molecular mechanisms of ciliary protein enrichment and its regulation among primary cilia and sensory cilia. Here we discuss the physiological demands on photoreceptors that have driven their evolution into neurons that rely on a highly specialized cilium for signaling changes in light intensity. We explore what is known and what is not known about how that specialization appears to have driven unique mechanisms for photoreceptor protein and membrane compartmentalization.
Collapse
Affiliation(s)
| | | | - Peter D. Calvert
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
7
|
Shi S, Ma K, Bin NR, Harada H, Xie X, Huang M, Liu H, Lee S, Wang XF, Adachi R, Monnier PP, Zhang L, Sugita S. Syntaxin-3 is dispensable for basal neurotransmission and synaptic plasticity in postsynaptic hippocampal CA1 neurons. Sci Rep 2020; 10:709. [PMID: 31959797 PMCID: PMC6971263 DOI: 10.1038/s41598-019-57388-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/30/2019] [Indexed: 01/17/2023] Open
Abstract
Recent evidence suggests that SNARE fusion machinery play critical roles in postsynaptic neurotransmitter receptor trafficking, which is essential for synaptic plasticity. However, the key SNAREs involved remain highly controversial; syntaxin-3 and syntaxin-4 are leading candidates for the syntaxin isoform underlying postsynaptic plasticity. In a previous study, we showed that pyramidal-neuron specific conditional knockout (cKO) of syntaxin-4 significantly reduces basal transmission, synaptic plasticity and impairs postsynaptic receptor trafficking. However, this does not exclude a role for syntaxin-3 in such processes. Here, we generated and analyzed syntaxin-3 cKO mice. Extracellular field recordings in hippocampal slices showed that syntaxin-3 cKO did not exhibit significant changes in CA1 basal neurotransmission or in paired-pulse ratios. Importantly, there were no observed differences during LTP in comparison to control mice. Syntaxin-3 cKO mice performed similarly as the controls in spatial and contextual learning tasks. Consistent with the minimal effects of syntaxin-3 cKO, syntaxin-3 mRNA level was very low in hippocampal and cortex pyramidal neurons, but strongly expressed in the corpus callosum and caudate axon fibers. Together, our data suggest that syntaxin-3 is dispensable for hippocampal basal neurotransmission and synaptic plasticity, and further supports the notion that syntaxin-4 is the major isoform mediating these processes.
Collapse
Affiliation(s)
- Shan Shi
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, 130021, China.,Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada
| | - Ke Ma
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, 130021, China. .,Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.
| | - Na-Ryum Bin
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Hidekiyo Harada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Ontario, M5T 2S8, Canada
| | - Xiaoyu Xie
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning, 116044, China
| | - Mengjia Huang
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Haiyu Liu
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Soomin Lee
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Xue Fan Wang
- Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Division of Genetics and Development, Krembil Research Institute, University Health Network, Ontario, M5T 2S8, Canada
| | - Roberto Adachi
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA
| | - Philippe P Monnier
- Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,Division of Genetics and Development, Krembil Research Institute, University Health Network, Ontario, M5T 2S8, Canada.,Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Liang Zhang
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada.,Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Shuzo Sugita
- Division of Fundamental Neurobiology, Krembil Research Institute, University Health Network, Toronto, Ontario, M5T 2S8, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
8
|
Frederick JM, Hanke-Gogokhia C, Ying G, Baehr W. Diffuse or hitch a ride: how photoreceptor lipidated proteins get from here to there. Biol Chem 2019; 401:573-584. [DOI: 10.1515/hsz-2019-0375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/02/2019] [Indexed: 12/23/2022]
Abstract
Abstract
Photoreceptors are polarized neurons, with specific subcellular compartmentalization and unique requirements for protein expression and trafficking. Each photoreceptor contains an outer segment (OS) where vision begins, an inner segment (IS) where protein synthesis occurs and a synaptic terminal for signal transmission to second-order neurons. The OS is a large, modified primary cilium attached to the IS by a slender connecting cilium (CC), the equivalent of the transition zone (TZ). Daily renewal of ~10% of the OS requires massive protein biosynthesis in the IS with reliable transport and targeting pathways. Transport of lipidated (‘sticky’) proteins depends on solubilization factors, phosphodiesterase δ (PDEδ) and uncoordinated protein-119 (UNC119), and the cargo dispensation factor (CDF), Arf-like protein 3-guanosine triphosphate (ARL3-GTP). As PDE6 and transducin still reside prominently in the OS of PDEδ and UNC119 germline knockout mice, respectively, we propose the existence of an alternate trafficking pathway, whereby lipidated proteins migrate in rhodopsin-containing vesicles of the secretory pathway.
Collapse
Affiliation(s)
- Jeanne M. Frederick
- Department of Ophthalmology and Visual Sciences , University of Utah Health Science Center , 65 Mario Capecchi Drive , Salt Lake City , UT 84132 , USA
| | - Christin Hanke-Gogokhia
- Department of Ophthalmology and Visual Sciences , University of Utah Health Science Center , 65 Mario Capecchi Drive , Salt Lake City , UT 84132 , USA
| | - Guoxin Ying
- Department of Ophthalmology and Visual Sciences , University of Utah Health Science Center , 65 Mario Capecchi Drive , Salt Lake City , UT 84132 , USA
| | - Wolfgang Baehr
- Department of Ophthalmology and Visual Sciences , University of Utah Health Science Center , 65 Mario Capecchi Drive , Salt Lake City , UT 84132 , USA
- Department of Neurobiology and Anatomy , University of Utah , Salt Lake City , UT 84112 , USA
- Department of Biology , University of Utah , Salt Lake City , UT 84132 , USA
| |
Collapse
|
9
|
Balancing the Photoreceptor Proteome: Proteostasis Network Therapeutics for Inherited Retinal Disease. Genes (Basel) 2019; 10:genes10080557. [PMID: 31344897 PMCID: PMC6722924 DOI: 10.3390/genes10080557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022] Open
Abstract
The light sensing outer segments of photoreceptors (PRs) are renewed every ten days due to their high photoactivity, especially of the cones during daytime vision. This demands a tremendous amount of energy, as well as a high turnover of their main biosynthetic compounds, membranes, and proteins. Therefore, a refined proteostasis network (PN), regulating the protein balance, is crucial for PR viability. In many inherited retinal diseases (IRDs) this balance is disrupted leading to protein accumulation in the inner segment and eventually the death of PRs. Various studies have been focusing on therapeutically targeting the different branches of the PR PN to restore the protein balance and ultimately to treat inherited blindness. This review first describes the different branches of the PN in detail. Subsequently, insights are provided on how therapeutic compounds directed against the different PN branches might slow down or even arrest the appalling, progressive blinding conditions. These insights are supported by findings of PN modulators in other research disciplines.
Collapse
|
10
|
De Loof A, Schoofs L. Flip-Flopping Retinal in Microbial Rhodopsins as a Template for a Farnesyl/Prenyl Flip-Flop Model in Eukaryote GPCRs. Front Neurosci 2019; 13:465. [PMID: 31133794 PMCID: PMC6515946 DOI: 10.3389/fnins.2019.00465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Thirty years after the first description and modeling of G protein coupled receptors (GPCRs), information about their mode of action is still limited. One of the questions that is hard to answer is: how do the allosteric changes in the GPCR induced by, e.g., ligand binding in the end activate a G protein-dependent intracellular pathway (e.g., via the cAMP or the phosphatidylinositol signal pathways). Another question relates to the role of prenylation of G proteins. Today's "consensus model" states that protein prenylation is required for the assembly of GPCR-G protein complexes. Although it is well-known that protein prenylation is the covalent addition of a farnesyl- or geranylgeranyl moiety to the C terminus of specific proteins, e.g., α or γ G protein, the reason for this strong covalent binding remains enigmatic. The arguments for a fundamental role for prenylation of G proteins other than just being a hydrophobic linker, are gradually accumulating. We uncovered a dilemma that at first glance may be considered physiologically irrelevant, however, it may cause a true change in paradigm. The consensus model suggests that the only functional role of prenylation is to link the G protein to the receptor. Does the isoprenoid nature of the prenyl group and its exact site of attachment somehow matter? Or, are there valid arguments favoring the alternative possibility that a key role of the G protein is to guide the covalently attached prenyl group to - and it hold it in - a very specific location in between specific helices of the receptor? Our model says that the farnesyl/prenyl group - aided by its covalent attachment to a G protein -might function in GPCRs as a horseshoe-shaped flexible (and perhaps flip-flopping) hydrophobic valve for restricting (though not fully inhibiting) the untimely passage of Ca2+, like retinal does for the passage of H+ in microbial rhodopsins that are ancestral to many GPCRs.
Collapse
Affiliation(s)
- Arnold De Loof
- Functional Genomics and Proteomics Group, Department of Biology, Zoological Institute, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
11
|
Identification of candidate biomarkers associated with apoptosis in melanosis coli: GNG5, LPAR3, MAPK8, and PSMC6. Biosci Rep 2019; 39:BSR20181369. [PMID: 30559147 PMCID: PMC6340944 DOI: 10.1042/bsr20181369] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose: Melanosis coli (MC) is a disorder of pigmentation of the wall of the colon, often identified at the time of colonoscopy. The aim of the present study is to identify candidate biomarkers for MC. Methods: The transcriptome data for MC (GSE78933) with five MC tissues and five corresponding normal tissues is obtained from the NCBI Gene Expression Omnibus (GEO) database. R/Bioconductor package limma was used to screen differently expressed genes (DEGs). ClueGO of cytoscape was applied for Gene Ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. Based on STRING V10 database, protein–protein interaction (PPI) network was constructed. The pathological tissue and normal tissue from 23 MC patients and 23 controls were collected, respectively. The relative expression of hub nodes was detected by qRT-PCR and Western blot. For regulating the expression of these genes, overexpression vector was constructed or siRNA transfection was used. Finally, apoptosis was detected by flow cytometry. Results: Total 1342 DEGs were screened, including 786 up-regulated and 556 down-regulated genes. These genes were mainly enriched in stimulatory C-type lectin receptor signaling pathway, polysaccharide biosynthetic process, intracellular, and oxidative phosphorylation. PPI network was then constructed with 426 DEGs and 895 interactions. Thereinto, G-protein subunit γ 5 (GNG5), lysophosphatidic acid receptor 3 (LPAR3), mitogen-activated protein kinase 8 (MAPK8), NHP2L1, proteasome 26S subunit, ATPase 6 (PSMC6), and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit β (PIK3CB) were hub nodes with higher degree. RT-PCR and Western blot results showed that GNG5, LPAR3, MAPK8, and PSMC6 were differently expressed with significance. The expression of these screened genes is also related with cell apoptosis. Conclusion: GNG5, LPAR3, MAPK8, and PSMC6 might be candidate biomarkers associated with apoptosis in MC.
Collapse
|
12
|
Sokolov M, Yadav RP, Brooks C, Artemyev NO. Chaperones and retinal disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:85-117. [PMID: 30635087 DOI: 10.1016/bs.apcsb.2018.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Defects in protein folding and trafficking are a common cause of photoreceptor degeneration, causing blindness. Photoreceptor cells present an unusual challenge to the protein folding and transport machinery due to the high rate of protein synthesis, trafficking and the renewal of the outer segment, a primary cilium that has been modified into a specialized light-sensing compartment. Phototransduction components, such as rhodopsin and cGMP-phosphodiesterase, and multimeric ciliary transport complexes, such as the BBSome, are hotspots for mutations that disrupt proteostasis and lead to the death of photoreceptors. In this chapter, we review recent studies that advance our understanding of the chaperone and transport machinery of phototransduction proteins.
Collapse
Affiliation(s)
- Maxim Sokolov
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
| | - Ravi P Yadav
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Celine Brooks
- Department of Ophthalmology, West Virginia University, Morgantown, WV, United States
| | - Nikolai O Artemyev
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, United States; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, United States.
| |
Collapse
|
13
|
Brooks C, Snoberger A, Belcastro M, Murphy J, Kisselev OG, Smith DM, Sokolov M. Archaeal Unfoldase Counteracts Protein Misfolding Retinopathy in Mice. J Neurosci 2018; 38:7248-7254. [PMID: 30012684 PMCID: PMC6096037 DOI: 10.1523/jneurosci.0905-18.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Accepted: 06/30/2018] [Indexed: 11/21/2022] Open
Abstract
Deregulation of cellular proteostasis due to the failure of the ubiquitin proteasome system to dispose of misfolded aggregation-prone proteins is a hallmark of various neurodegenerative diseases in humans. Microorganisms have evolved to survive massive protein misfolding and aggregation triggered by heat shock using their protein-unfolding ATPases (unfoldases) from the Hsp100 family. Because the Hsp100 chaperones are absent in homoeothermic mammals, we hypothesized that the vulnerability of mammalian neurons to misfolded proteins could be mitigated by expressing a xenogeneic unfoldase. To test this idea, we expressed proteasome-activating nucleotidase (PAN), a protein-unfolding ATPase from thermophilic Archaea, which is homologous to the 19S eukaryotic proteasome and similar to the Hsp100 family chaperones in rod photoreceptors of mice. We found that PAN had no obvious effect in healthy rods; however, it effectively counteracted protein-misfolding retinopathy in Gγ1 knock-out mice. We conclude that archaeal PAN can rescue a protein-misfolding neurodegenerative disease, likely by recognizing misfolded mammalian proteins.SIGNIFICANCE STATEMENT This study demonstrates successful therapeutic application of an archaeal molecular chaperone in an animal model of neurodegenerative disease. Introducing the archaeal protein-unfolding ATPase proteasome-activating nucleotidase (PAN) into the retinal photoreceptors of mice protected these neurons from the cytotoxic effect of misfolded proteins. We propose that xenogeneic protein-unfolding chaperones could be equally effective against other types of neurodegenerative diseases of protein-misfolding etiology.
Collapse
Affiliation(s)
| | - Aaron Snoberger
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, and
| | | | | | - Oleg G Kisselev
- Department of Ophthalmology, St. Louis University, St. Louis, Missouri 63104
| | - David M Smith
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, and
| | - Maxim Sokolov
- Department of Ophthalmology,
- Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, and
| |
Collapse
|