1
|
Kajimura Y, Dong S, Tessari A, Orlacchio A, Thoms A, Cufaro MC, Di Marco F, Amari F, Chen M, Soliman SHA, Rizzotto L, Zhang L, Sunilkumar D, Amann JM, Carbone DP, Ahmed A, Fiermonte G, Freitas MA, Lodi A, Del Boccio P, Tessarollo L, Palmieri D, Coppola V. An in vivo "turning model" reveals new RanBP9 interactions in lung macrophages. Cell Death Discov 2025; 11:171. [PMID: 40223093 PMCID: PMC11994786 DOI: 10.1038/s41420-025-02456-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 03/13/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
The biological functions of the scaffold protein Ran Binding Protein 9 (RanBP9) remain elusive in macrophages or any other cell type where this protein is expressed together with its CTLH (C-terminal to LisH) complex partners. We have engineered a new mouse model, named RanBP9-TurnX, where RanBP9 fused to three copies of the HA tag (RanBP9-3xHA) can be turned into RanBP9-V5 tagged upon Cre-mediated recombination. We created this model to enable stringent biochemical studies at cell type specific level throughout the entire organism. Here, we have used this tool crossed with LysM-Cre transgenic mice to identify RanBP9 interactions in lung macrophages. We show that RanBP9-V5 and RanBP9-3xHA can be both co-immunoprecipitated with the known members of the CTLH complex from the same whole lung lysates. However, more than ninety percent of the proteins pulled down by RanBP9-V5 differ from those pulled-down by RanBP9-HA. The lung RanBP9-V5 associated proteome includes previously unknown interactions with macrophage-specific proteins as well as with players of the innate immune response, DNA damage response, metabolism, and mitochondrial function. This work provides the first lung specific RanBP9-associated interactome in physiological conditions and reveals that RanBP9 and the CTLH complex could be key regulators of macrophage bioenergetics and immune functions.
Collapse
Affiliation(s)
- Yasuko Kajimura
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Division of Hematology, Diabetes, Metabolism and Endocrinology, Yamaguchi University Hospital, Yamaguchi, Japan
| | - Shuxin Dong
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Anna Tessari
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Oncology Unit, AULSS 5 Polesana, Rovigo, Italy
| | - Arturo Orlacchio
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Alexandra Thoms
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Pelotonia Summer Fellow, Kenyon College, CAMELOT Program, Gambier, OH, USA
| | - Maria Concetta Cufaro
- Analytical Biochemistry and Proteomics Research Unit, CAST (Center for Advanced Studies and Technology), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Federica Di Marco
- Analytical Biochemistry and Proteomics Research Unit, CAST (Center for Advanced Studies and Technology), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Min Chen
- Genetically Engineered Mouse Modeling Core, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Shimaa H A Soliman
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Simpson Querrey Institute for Epigenetics, Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Lara Rizzotto
- Gene Editing Shared Resource, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Liwen Zhang
- Proteomic Shared Resource, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Damu Sunilkumar
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Joseph M Amann
- Division of Medical Oncology, Ohio State Wexner Medical Center, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - David P Carbone
- Division of Medical Oncology, Ohio State Wexner Medical Center, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Amer Ahmed
- Department of Biosciences, Biotechnology and Environment, University of Bari, 70125, Bari, Italy
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnology and Environment, University of Bari, 70125, Bari, Italy
| | - Mike A Freitas
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Proteomic Shared Resource, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX, 78723, USA
| | - Piero Del Boccio
- Analytical Biochemistry and Proteomics Research Unit, CAST (Center for Advanced Studies and Technology), University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Lino Tessarollo
- Neural Development Section, Mouse Cancer Genetics Program, NCI/Center for Cancer Research, NIH, Frederick, MD, 21702, USA
| | - Dario Palmieri
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Gene Editing Shared Resource, The Ohio State University and Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University Arthur G. James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Kajimura Y, Tessari A, Orlacchio A, Thoms A, Cufaro MC, Marco FD, Amari F, Chen M, Soliman SHA, Rizzotto L, Zhang L, Amann J, Carbone DP, Ahmed A, Fiermonte G, Freitas M, Lodi A, Boccio PD, Palmieri D, Coppola V. An in vivo "turning model" reveals new RanBP9 interactions in lung macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595416. [PMID: 38826292 PMCID: PMC11142189 DOI: 10.1101/2024.05.22.595416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The biological functions of the scaffold protein Ran Binding Protein 9 (RanBP9) remain elusive in macrophages or any other cell type where this protein is expressed together with its CTLH (C-terminal to LisH) complex partners. We have engineered a new mouse model, named RanBP9-TurnX, where RanBP9 fused to three copies of the HA tag (RanBP9-3xHA) can be turned into RanBP9-V5 tagged upon Cre-mediated recombination. We created this model to enable stringent biochemical studies at cell type specific level throughout the entire organism. Here, we have used this tool crossed with LysM-Cre transgenic mice to identify RanBP9 interactions in lung macrophages. We show that RanBP9-V5 and RanBP9-3xHA can be both co-immunoprecipitated with the known members of the CTLH complex from the same whole lung lysates. However, more than ninety percent of the proteins pulled down by RanBP9-V5 differ from those pulled-down by RanBP9-HA. The lung RanBP9-V5 associated proteome includes previously unknown interactions with macrophage-specific proteins as well as with players of the innate immune response, DNA damage response, metabolism, and mitochondrial function. This work provides the first lung specific RanBP9-associated interactome in physiological conditions and reveals that RanBP9 and the CTLH complex could be key regulators of macrophage bioenergetics and immune functions.
Collapse
|
3
|
Walker CK, Greathouse KM, Liu E, Muhammad HM, Boros BD, Freeman CD, Seo JV, Herskowitz JH. Comparison of Golgi-Cox and Intracellular Loading of Lucifer Yellow for Dendritic Spine Density and Morphology Analysis in the Mouse Brain. Neuroscience 2022; 498:1-18. [PMID: 35752428 PMCID: PMC9420811 DOI: 10.1016/j.neuroscience.2022.06.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 10/17/2022]
Abstract
Dendritic spines are small protrusions on dendrites that serve as the postsynaptic site of the majority of excitatory synapses. These structures are important for normal synaptic transmission, and alterations in their density and morphology have been documented in various disease states. Over 130 years ago, Ramón y Cajal used Golgi-stained tissue sections to study dendritic morphology. Despite the array of technological advances, including iontophoretic microinjection of Lucifer yellow (LY) fluorescent dye, Golgi staining continues to be one of the most popular approaches to visualize dendritic spines. Here, we compared dendritic spine density and morphology among pyramidal neurons in layers 2/3 of the mouse medial prefrontal cortex (mPFC) and pyramidal neurons in hippocampal CA1 using three-dimensional digital reconstructions of (1) brightfield microscopy z-stacks of Golgi-impregnated dendrites and (2) confocal microscopy z-stacks of LY-filled dendrites. Analysis of spine density revealed that the LY microinjection approach enabled detection of approximately three times as many spines as the Golgi staining approach in both brain regions. Spine volume measurements were larger using Golgi staining compared to LY microinjection in both mPFC and CA1. Spine length was mostly comparable between techniques in both regions. In the mPFC, head diameter was similar for Golgi staining and LY microinjection. However, in CA1, head diameter was approximately 50% smaller on LY-filled dendrites compared to Golgi staining. These results indicate that Golgi staining and LY microinjection yield different spine density and morphology measurements, with Golgi staining failing to detect dendritic spines and overestimating spine size.
Collapse
Affiliation(s)
- Courtney K Walker
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Kelsey M Greathouse
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Evan Liu
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Hamad M Muhammad
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Benjamin D Boros
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Cameron D Freeman
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Jung Vin Seo
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA
| | - Jeremy H Herskowitz
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, USA.
| |
Collapse
|
4
|
Wang H, Muthu Karuppan MK, Nair M, Lakshmana MK. Autophagy-Dependent Increased ADAM10 Mature Protein Induced by TFEB Overexpression Is Mediated Through PPARα. Mol Neurobiol 2021; 58:2269-2283. [PMID: 33417226 DOI: 10.1007/s12035-020-02230-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Nonamyloidogenic processing of amyloid precursor protein (APP) by augmenting ADAM10 is a promising therapeutic strategy for Alzheimer's disease (AD). Therefore identification of molecular pathways that regulate ADAM10 expression is crucial. Autophagy is strongly dysregulated in AD, and TFEB was recently shown to be a master regulator of autophagy-lysosome pathway (ALP). Here, we report that TFEB expression in HeLa cells increased ADAM10 mature form by 72% (p < 0.01, n = 4), while TFEB knockdown by CRISPR strategy reduced ADAM10 mature form by 36% (p < 0.05, n = 4). Autophagy inhibition by 3-methyladenine (3-MA), but not bafilomycin A1 (BAF1), reduced ADAM10 mature form by 49% (p < 0.05, n = 4) in the TFEB expressing HeLa cells. Autophagy activation by 3 h of starvation increased ADAM10 to 91% (p < 0.001, n = 6) relative to 51% (p < 0.01, n = 6) in the nutrient-fed cells. Further, siRNAs targeted against PPARα in HeLa cells decreased ADAM10 levels by 28% (p < 0.05, n = 6) relative to the cells treated with scrambled siRNAs. Further, incubation of EGFP-TFEB expressing HeLa cells with PPARα antagonist, but not PPARβ or PPARγ antagonists, prevented TFEB-induced increase in ADAM10 levels. Importantly, flag-TFEB expression in the brain also increased ADAM10 by 60% (p < 0.05, n = 3) in the cortical and 34% (p < 0.001, n = 3) in the hippocampal homogenates. ADAM10 activity also increased by 57% (p < 0.01, n = 3) in the HeLa cells. Finally, TFEB-induced ADAM10 potentiation led to increased secretion of sAPPα by 154% (p < 0.001, n = 3) in the cortex and 62% (p < 0.001, n = 3) in the hippocampus. Thus, TFEB expression enhances nonamyloidogenic processing of APP. In conclusion, TFEB expression induces ADAM10 in an autophagy-dependent manner through PPARα.
Collapse
Affiliation(s)
- Hongjie Wang
- Institute for Human Health & Disease Intervention (I-HEALTH), Department of Chemistry and Biochemistry, Center for Molecular Biology and Biotechnology, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Mohan Kumar Muthu Karuppan
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA
| | - Madepalli K Lakshmana
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200, 8th Street, University Park, Miami, FL, 33199, USA.
| |
Collapse
|
5
|
Kang DE, Woo JA. Cofilin, a Master Node Regulating Cytoskeletal Pathogenesis in Alzheimer's Disease. J Alzheimers Dis 2020; 72:S131-S144. [PMID: 31594228 PMCID: PMC6971827 DOI: 10.3233/jad-190585] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The defining pathological hallmarks of Alzheimer’s disease (AD) are proteinopathies marked by the amyloid-β (Aβ) peptide and hyperphosphorylated tau. In addition, Hirano bodies and cofilin-actin rods are extensively found in AD brains, both of which are associated with the actin cytoskeleton. The actin-binding protein cofilin known for its actin filament severing, depolymerizing, nucleating, and bundling activities has emerged as a significant player in AD pathogenesis. In this review, we discuss the regulation of cofilin by multiple signaling events impinging on LIM kinase-1 (LIMK1) and/or Slingshot homolog-1 (SSH1) downstream of Aβ. Such pathophysiological signaling pathways impact actin dynamics to regulate synaptic integrity, mitochondrial translocation of cofilin to promote neurotoxicity, and formation of cofilin-actin pathology. Other intracellular signaling proteins, such as β-arrestin, RanBP9, Chronophin, PLD1, and 14-3-3 also impinge on the regulation of cofilin downstream of Aβ. Finally, we discuss the role of activated cofilin as a bridge between actin and microtubule dynamics by displacing tau from microtubules, thereby destabilizing tau-induced microtubule assembly, missorting tau, and promoting tauopathy.
Collapse
Affiliation(s)
- David E Kang
- Byrd Institute and Alzheimer's Center, USF Health Morsani College of Medicine, Tampa, FL, USA.,Department of Molecular Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA.,Division of Research, James A. Haley VA Hospital, Tampa, FL, USA
| | - Jung A Woo
- Byrd Institute and Alzheimer's Center, USF Health Morsani College of Medicine, Tampa, FL, USA.,Department of Molecular Pharmacology and Physiology, USF Health Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
6
|
Margabandhu G, Vanisree AJ. Dopamine, a key factor of mitochondrial damage and neuronal toxicity on rotenone exposure and also parkinsonic motor dysfunction-Impact of asiaticoside with a probable vesicular involvement. J Chem Neuroanat 2020; 106:101788. [PMID: 32278634 DOI: 10.1016/j.jchemneu.2020.101788] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/16/2020] [Accepted: 03/19/2020] [Indexed: 12/18/2022]
Abstract
Persuasive evidence propose that the toxicity of dopamine in parkinsonism and the loss of dopaminergic neurons are the earliest events during the pathogenesis of Parkinson's disease (PD). In our earlier study, Asiaticoside (AS), a triterpenoid saponin isolated from Centella asiatica was shown to exert a neuroprotective effect against hemiparkinsonism, purportedly due to phosphoinositides (PI)-assisted cytodynamics and synaptic function. Here, we evaluate AS in the modulation of dopamine (DA), mitochondrial integrity and neurite variations in vitro and motor dysfunctions in vivo. PC12 cells challenged with rotenone-(ROT) (0.1 μM/mL) were exposed to AS and l-DOPA (10 mM and 20 μM/mL respectively). The protein expressions of Bax and Bcl-2 that regulate cell death were assessed following neurite length assays. Rats were distributed into 6 groups (6 rats/group): Sham, Vehicle controls, ROT-infused (6 μg/μl/kg), AS- treated (50 mg/kg/day), Drug control, and ROT + L-DOPA-treated (6 mg/kg/day) groups. At the end of the experimental period, the rats were sacrificed after performing motor behavioral analysis, and the striatum was dissected out. The contents of synaptic vesicular and cytosolic DA were analyzed. Further, the levels of striatal PI were also measured. ROT had caused significant reduction in the neurite outgrowth in the exposed PC12 cells while the tested concentrations of AS and l-DOPA can exert their protective effect on the stunted neurite growth. The levels of Bax, Bcl-2, and cytochrome c which were significantly disturbed by ROT, could also be affected by AS thereby suggesting its effect on neurons. AS treatment caused an improved motor performance, vesicular and cytosolic DA, and striatal PI. These pre-clinical findings force us to speculate that AS could be a potential drug candidate in combating ROT-induced variations that are possibly precipitated by varied vesicular trafficking of DA.
Collapse
Affiliation(s)
- Gopi Margabandhu
- Unit of Molecular Neurobiology, Department of Biochemistry, University of Madras, Guindy Campus, Chennai, 600 025, Tamilnadu, India.
| | - Arambakkam Janardhanam Vanisree
- Unit of Molecular Neurobiology, Department of Biochemistry, University of Madras, Guindy Campus, Chennai, 600 025, Tamilnadu, India.
| |
Collapse
|
7
|
Qin C, Zhang Q, Wu G. RANBP9 suppresses tumor proliferation in colorectal cancer. Oncol Lett 2019; 17:4409-4416. [PMID: 30988811 PMCID: PMC6447939 DOI: 10.3892/ol.2019.10134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
RAN binding protein 9 (RANBP9) is widely expressed in mammalian tissues, including osteosarcoma, lung, gastric and breast cancer tissues. However, currently, not much is known about the role of RANBP9 in colorectal cancer (CRC). In the present study, RANBP9 expression in CRC tissues and cell lines was measured by immunohistochemistry and western blotting, respectively. Subsequently, RANBP9-short hairpin RNA (shRNA) and RANBP9 plasmids were constructed and transfected into HCT116 and HT29 cells. The effects of RANBP9 knockdown were assessed by Cell Counting kit-8 and colony formation assays, and its effects on tumorigenicity in a nude mouse animal model were investigated. The effect of RANBP9-shRNA on cell cycle progression was analyzed by flow cytometry, while cell cycle-associated protein expression levels were examined by western blotting. Compared with in paired normal mucosa, RANBP9 was overexpressed in CRC tissues. Inhibition of RANBP9 in HCT116 and HT29 cells significantly promoted cell growth, colony formation and S phase transition, and increased tumorigenesis in vivo. Accordingly, RANBP9 overexpression inhibited cell growth and colony formation. Knockdown of RANBP9 was associated with upregulated cyclin A2 in the two cell lines. In conclusion, RANBP9 served an inhibitory role in CRC in vitro and in vivo. Therefore, RANBP9 may be considered a potential target for treatment of CRC.
Collapse
Affiliation(s)
- Chunzhi Qin
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Qin Zhang
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Guangbin Wu
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
8
|
Salemi LM, Maitland MER, McTavish CJ, Schild-Poulter C. Cell signalling pathway regulation by RanBPM: molecular insights and disease implications. Open Biol 2018; 7:rsob.170081. [PMID: 28659384 PMCID: PMC5493780 DOI: 10.1098/rsob.170081] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/01/2017] [Indexed: 12/25/2022] Open
Abstract
RanBPM (Ran-binding protein M, also called RanBP9) is an evolutionarily conserved, ubiquitous protein which localizes to both nucleus and cytoplasm. RanBPM has been implicated in the regulation of a number of signalling pathways to regulate several cellular processes such as apoptosis, cell adhesion, migration as well as transcription, and plays a critical role during development. In addition, RanBPM has been shown to regulate pathways implicated in cancer and Alzheimer's disease, implying that RanBPM has important functions in both normal and pathological development. While its functions in these processes are still poorly understood, RanBPM has been identified as a component of a large complex, termed the CTLH (C-terminal to LisH) complex. The yeast homologue of this complex functions as an E3 ubiquitin ligase that targets enzymes of the gluconeogenesis pathway. While the CTLH complex E3 ubiquitin ligase activity and substrates still remain to be characterized, the high level of conservation between the complexes in yeast and mammals infers that the CTLH complex could also serve to promote the degradation of specific substrates through ubiquitination, therefore suggesting the possibility that RanBPM's various functions may be mediated through the activity of the CTLH complex.
Collapse
Affiliation(s)
- Louisa M Salemi
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Matthew E R Maitland
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Christina J McTavish
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| | - Caroline Schild-Poulter
- Robarts Research Institute, Department of Biochemistry, Schulich School of Medicine and Dentistry, The University of Western Ontario, 1151 Richmond Street North, London, Ontario, Canada N6A 5B7
| |
Collapse
|
9
|
Das S, Suresh B, Kim HH, Ramakrishna S. RanBPM: a potential therapeutic target for modulating diverse physiological disorders. Drug Discov Today 2017; 22:1816-1824. [PMID: 28847759 DOI: 10.1016/j.drudis.2017.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 06/26/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023]
Abstract
The Ran-binding protein microtubule-organizing center (RanBPM) is a highly conserved nucleocytoplasmic protein involved in a variety of intracellular signaling pathways that control diverse cellular functions. RanBPM interacts with proteins that are linked to various diseases, including Alzheimer's disease (AD), schizophrenia (SCZ), and cancer. In this article, we define the characteristics of the scaffolding protein RanBPM and focus on its interaction partners in diverse physiological disorders, such as neurological diseases, fertility disorders, and cancer.
Collapse
Affiliation(s)
- Soumyadip Das
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Bharathi Suresh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Hyongbum Henry Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, South Korea; Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, South Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
10
|
Qiao R, Li S, Zhou M, Chen P, Liu Z, Tang M, Zhou J. In-depth analysis of the synaptic plasma membrane proteome of small hippocampal slices using an integrated approach. Neuroscience 2017; 353:119-132. [PMID: 28435053 DOI: 10.1016/j.neuroscience.2017.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/24/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
Comprehensive knowledge of the synaptic plasma membrane (SPM) proteome of a distinct brain region in a defined pathological state would greatly advance the understanding of the underlying biology of synaptic plasticity. The development of innovative approaches for studying the SPM proteome of small brain tissues is highly desired. This study presents a suitable protocol that integrates biotinylation-based affinity capture of cell surface-exposed proteins, isolation of synaptosomes, and biochemical extraction of SPM proteins from biotinylated hippocampal slices. The effectiveness of this integrated method was initially confirmed using immunoblot analysis of synaptic markers. Subsequently, we used highly sensitive mass spectrometry and streamlined bioinformatics to analyze the obtained SPM protein-enriched fraction. Our workflow positively identified 241 SPM proteins comprising 85 previously reported classical proteins from the pre- and/or post-synaptic membrane and 156 nonclassical proteins that localized to both the plasma membrane and synapse, and have not been previously reported as SPM proteins. Further analyses revealed considerable similarities in the physicochemical and functional properties of these proteins. Analysis of the interaction network using STRING indicated that the two groups showed a relatively strong functional correlation. Using MCODE analysis, we observed that 65 nonclassical SPM proteins formed 12 highly interconnected clusters with 47 classical SPM proteins, suggesting that they were the more likely SPM candidates. Taken together, the results of this study provide an integrated tool for analyzing the SPM proteome of small brain tissues, as well as a dataset of putative novel SPM proteins to improve the understanding of hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Rui Qiao
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Shuiming Li
- Shenzhen Key Laboratory of Microbiology and Gene Engineering, Shenzhen University, Shenzhen 518060, China
| | - Mi Zhou
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Penghui Chen
- Department of Neurobiology, The Third Military Medical University, Chongqing 400038, China
| | - Zhao Liu
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Min Tang
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China
| | - Jian Zhou
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China.
| |
Collapse
|
11
|
Ma J, Duan Y, Qin Z, Wang J, Liu W, Xu M, Zhou S, Cao X. Overexpression of αCaMKII impairs behavioral flexibility and NMDAR-dependent long-term depression in the medial prefrontal cortex. Neuroscience 2015; 310:528-40. [PMID: 26415772 DOI: 10.1016/j.neuroscience.2015.09.051] [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: 06/24/2015] [Revised: 09/02/2015] [Accepted: 09/20/2015] [Indexed: 01/24/2023]
Abstract
The medial prefrontal cortex (mPFC) participates in the behavioral flexibility. As a major downstream molecule in the NMDA receptor signaling, alpha-Ca(2+)/calmodulin-dependent protein kinase II (αCaMKII) is crucial for hippocampal long-term potentiation (LTP) and hippocampus-related memory. However, the role of αCaMKII in mPFC-related behavioral flexibility and mPFC synaptic plasticity remains elusive. In the present study, using chemical-genetic approaches to temporally up-regulate αCaMKII activity, we found that αCaMKII-F89G transgenic mice exhibited impaired behavioral flexibility in Y-water maze arm reversal task. Notably, in vitro electrophysiological analysis showed normal basal synaptic transmission, LTP and depotentiation, but selectively impaired NMDAR-dependent long-term depression (LTD) in the mPFC of αCaMKII-F89G transgenic mice. In accordance with the deficit in NMDAR-dependent LTD, αCaMKII-F89G transgenic mice exhibited impaired AMPAR internalization during NMDAR-dependent chemical LTD expression in the mPFC. Furthermore, the above deficits in behavioral flexibility, NMDAR-dependent LTD and AMPAR internalization could all be reversed by 1-naphthylmethyl (NM)-PP1, a specific inhibitor of exogenous αCaMKII-F89G activity. Taken together, our results for the first time indicate that αCaMKII overexpression in the forebrain impairs behavioral flexibility and NMDAR-dependent LTD in the mPFC, and supports the notion that there is a close relationship between NMDAR-dependent LTD and behavioral flexibility.
Collapse
Affiliation(s)
- J Ma
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Y Duan
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Z Qin
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - J Wang
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - W Liu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - M Xu
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - S Zhou
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - X Cao
- Key Laboratory of Brain Functional Genomics (East China Normal University), Ministry of Education, Shanghai Key Laboratory of Brain Functional Genomics (East China Normal University), School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| |
Collapse
|
12
|
Wang R, Wang H, Carrera I, Xu S, Lakshmana MK. COPS5 protein overexpression increases amyloid plaque burden, decreases spinophilin-immunoreactive puncta, and exacerbates learning and memory deficits in the mouse brain. J Biol Chem 2015; 290:9299-309. [PMID: 25713139 DOI: 10.1074/jbc.m114.595926] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 01/05/2023] Open
Abstract
Brain accumulation of neurotoxic amyloid β (Aβ) peptide because of increased processing of amyloid precursor protein (APP), resulting in loss of synapses and neurodegeneration, is central to the pathogenesis of Alzheimer disease (AD). Therefore, the identification of molecules that regulate Aβ generation and those that cause synaptic damage is crucial for future therapeutic approaches for AD. We demonstrated previously that COPS5 regulates Aβ generation in neuronal cell lines in a RanBP9-dependent manner. Consistent with the data from cell lines, even by 6 months, COPS5 overexpression in APΔE9 mice (APΔE9/COPS5-Tg) significantly increased Aβ40 levels by 32% (p < 0.01) in the cortex and by 28% (p < 0.01) in the hippocampus, whereas the increases for Aβ42 were 37% (p < 0.05) and 34% (p < 0.05), respectively. By 12 months, the increase was even more robust. Aβ40 levels increased by 63% (p < 0.001) in the cortex and by 65% (p < 0.001) in the hippocampus. Similarly, Aβ42 levels were increased by 69% (p < 0.001) in the cortex and by 71% (p < 0.011) in the hippocampus. Increased Aβ levels were translated into an increased amyloid plaque burden both in the cortex (54%, p < 0.01) and hippocampus (64%, p < 0.01). Interestingly, COPS5 overexpression increased RanBP9 levels in the brain, which, in turn, led to increased amyloidogenic processing of APP, as reflected by increased levels of sAPPβ and decreased levels of sAPPα. Furthermore, COPS5 overexpression reduced spinophilin in both the cortex (19%, p < 0.05) and the hippocampus (20%, p < 0.05), leading to significant deficits in learning and memory skills. Therefore, like RanBP9, COPS5 also plays a pivotal role in amyloid pathology in vivo.
Collapse
Affiliation(s)
- Ruizhi Wang
- From the Section of Neurobiology, Torrey Pines Institute for Molecular Studies, Port Saint Lucie, Florida 34987
| | - Hongjie Wang
- From the Section of Neurobiology, Torrey Pines Institute for Molecular Studies, Port Saint Lucie, Florida 34987
| | - Ivan Carrera
- the Department of Neuroscience, Euroespes Biotechnology, Poligono de Bergondo, Nave F, 15165A, A Coruna, Spain, and
| | - Shaohua Xu
- the Florida Institute of Technology, Melbourne, Florida 32901
| | - Madepalli K Lakshmana
- From the Section of Neurobiology, Torrey Pines Institute for Molecular Studies, Port Saint Lucie, Florida 34987,
| |
Collapse
|
13
|
Appraisal of the effect of brain impregnation duration on neuronal staining and morphology in a modified Golgi-Cox method. J Neurosci Methods 2014; 235:193-207. [PMID: 25063423 DOI: 10.1016/j.jneumeth.2014.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/12/2014] [Accepted: 07/15/2014] [Indexed: 11/20/2022]
Abstract
BACKGROUND Golgi-Cox staining method is considered as one of the best neurohistological and fascinating staining techniques to reveal the cytoarchitecture of the brain. Requirement of longer time (more than a month), laborious section processing steps, requirement of sophisticated equipment's and costly ready to use kits limits extensive use of this technique. NEW METHOD The need for a modified staining technique is to overcome some of these hurdles. Here we describe a modification of Golgi-Cox staining involving reduced impregnation time (7 days), omitting tissue dehydration steps, and alterations in section processing steps. Different impregnation duration (7 days, 14 days, 1 month, 6 months and 10 months) effects on optimized staining of dorsal hippocampus and basolateral amygdala were investigated. RESULTS Modified Golgi-Cox staining method was found to be effective in staining rat hippocampus and amygdala. Impregnation for 7 days, 14 days and 1 month resulted in giving good results and they were comparable. However, artifacts were slightly elevated with 6 months group but not extensively. Impregnation for 10 months negatively affected the staining process. COMPARISON WITH EXISTING METHOD(S) Compared to existing methods the current method was found to be cost effective, fast, reliable and can be executed in labs where infrastructure is limited. CONCLUSIONS Current modification considerably benefitted in obtaining better results (good clarity and lesser artifact) in a short time. Longer impregnated brain sections were found to be unsuitable for morphometric evaluation due to more stain precipitation and artifact. The modified technique can be used to study cellular architecture in other brain regions.
Collapse
|