24 results on '"dynamin-1"'
Search Results
2. Dynamin-1 is a potential mediator in cancer-related cognitive impairment
- Author
-
Ng, Ding Quan, Hudson, Casey, Nguyen, Tracy, Gupta, Sukesh Kumar, Koh, Yong Qin, Acharya, Munjal M, and Chan, Alexandre
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Mental Health ,Neurodegenerative ,Breast Cancer ,Behavioral and Social Science ,Acquired Cognitive Impairment ,Women's Health ,Cancer ,Pediatric ,Basic Behavioral and Social Science ,Neurosciences ,Dementia ,Aging ,Brain Disorders ,2.1 Biological and endogenous factors ,Cancer-related cognitive impairment ,Chemobrain ,Dynamin-1 ,Extracellular vesicle ,Exosome ,Pharmacology and Pharmaceutical Sciences ,Public Health and Health Services ,Neurology & Neurosurgery ,Pharmacology and pharmaceutical sciences ,Biological psychology - Abstract
Dynamin-1 (DNM1) is crucial for synaptic activity, neurotransmission, and associative memory, positioning it as a potential biomarker of cancer-related cognitive impairment (CRCI), a neurological consequence of cancer treatment characterized by memory loss, poor concentration, and impaired executive function. Through a stepwise approach, this study investigated the role of DNM1 in CRCI pathogenesis, incorporating both human data and animal models. The human study recruited newly diagnosed, chemotherapy-naïve adolescent and young adult cancer and non-cancer controls to complete a cognitive instrument (FACT-Cog) and blood draws for up to three time points. Following that, a syngeneic young-adult WT (C57BL/6) female mouse model of breast cancer chemobrain was developed to study DNM1 expression in the hippocampus. Samples from eighty-six participants with 30 adolescent and young adult (AYA) cancer and 56 non-cancer participants were analyzed. DNM1 levels were 32 % lower (P = 0.041) among cancer participants compared to non-cancer prior to treatment. After receiving cytotoxic treatment, cognitively impaired cancer patients were found to have 46 % lower DNM1 levels than those without impairment (P = 0.049). In murine breast cancer-bearing mice receiving chemotherapy, we found a greater than 40 % decline (P
- Published
- 2024
3. Cancer-related cognitive impairment and hippocampal functioning: The role of dynamin-1
- Author
-
Nordhjem, Barbara Johanne Thomas and Hjalgrim, Lisa Lyngsie
- Published
- 2025
- Full Text
- View/download PDF
4. A recurrent de novo splice site variant involving DNM1 exon 10a causes developmental and epileptic encephalopathy through a dominant-negative mechanism.
- Author
-
Parthasarathy, Shridhar, Ruggiero, Sarah McKeown, Gelot, Antoinette, Soardi, Fernanda C, Ribeiro, Bethânia F R, Pires, Douglas E V, Ascher, David B, Schmitt, Alain, Rambaud, Caroline, Represa, Alfonso, Xie, Hongbo M, Lusk, Laina, Wilmarth, Olivia, McDonnell, Pamela Pojomovsky, Juarez, Olivia A, Grace, Alexandra N, Buratti, Julien, Mignot, Cyril, Gras, Domitille, and Nava, Caroline
- Subjects
- *
BRAIN diseases , *GENETIC variation , *SYNAPTIC vesicles , *PEOPLE with epilepsy , *MISSENSE mutation - Abstract
Heterozygous pathogenic variants in DNM1 cause developmental and epileptic encephalopathy (DEE) as a result of a dominant-negative mechanism impeding vesicular fission. Thus far, pathogenic variants in DNM1 have been studied with a canonical transcript that includes the alternatively spliced exon 10b. However, after performing RNA sequencing in 39 pediatric brain samples, we find the primary transcript expressed in the brain includes the downstream exon 10a instead. Using this information, we evaluated genotype-phenotype correlations of variants affecting exon 10a and identified a cohort of eleven previously unreported individuals. Eight individuals harbor a recurrent de novo splice site variant, c.1197−8G>A (GenBank: NM_001288739.1), which affects exon 10a and leads to DEE consistent with the classical DNM1 phenotype. We find this splice site variant leads to disease through an unexpected dominant-negative mechanism. Functional testing reveals an in-frame upstream splice acceptor causing insertion of two amino acids predicted to impair oligomerization-dependent activity. This is supported by neuropathological samples showing accumulation of enlarged synaptic vesicles adherent to the plasma membrane consistent with impaired vesicular fission. Two additional individuals with missense variants affecting exon 10a, p.Arg399Trp and p.Gly401Asp, had a similar DEE phenotype. In contrast, one individual with a missense variant affecting exon 10b, p.Pro405Leu, which is less expressed in the brain, had a correspondingly less severe presentation. Thus, we implicate variants affecting exon 10a as causing the severe DEE typically associated with DNM1 -related disorders. We highlight the importance of considering relevant isoforms for disease-causing variants as well as the possibility of splice site variants acting through a dominant-negative mechanism. Heterozygous pathogenic variants in DNM1 cause developmental and epileptic encephalopathy. Here, we perform RNA sequencing in brain samples and find non-canonical, alternatively spliced transcripts are the most abundant. We highlight the importance of considering relevant isoforms for variant interpretation and the possibility of intronic variants causing disease through dominant-negative mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. Lipopolysaccharide Accelerates Neuropilin-1 Protein Degradation by Activating the Large GTPase Dynamin-1 in Macrophages.
- Author
-
Wu, Suhua, Huang, Yueling, Huang, Xinliang, and Dai, Xiaoyan
- Subjects
- *
PROTEOLYSIS , *GUANOSINE triphosphatase , *LIPOPOLYSACCHARIDES , *TYPE 2 diabetes , *MACROPHAGES , *MACROPHAGE activation syndrome - Abstract
Neuropilin-1 (Nrp1) is highly expressed in macrophages and plays a critical role in acute and chronic inflammation-associated diseases, such as sepsis, type II diabetes, and metabolic syndrome. Therefore, it is of importance to understand the regulation of Nrp1. It is known that lipopolysaccharide (LPS) downregulates Nrp1 mRNA levels through the NF-κB signaling in macrophages. However, whether and how LPS regulates Nrp1 protein degradation remain unknown. Here, we show that LPS promotes Nrp1 protein decay through a lysosome-dependent manner. Liver kinase B1 (LKB1)-Rab7 does not mediate this process. However, the large GTPase dynamin-1 (Dyn1) but not Dyn2 is involved in LPS-accelerated Nrp1 degradation. Mechanistically, LPS activates Dyn1 by attenuating p-Dyn1 (Ser774) levels, implying increased Nrp1 endocytosis and consequent degradation. As a result, blocking Nrp1 degradation by Dyn1 siRNA attenuates LPS-induced inflammatory response. Collectively, our study shows that LPS promotes Nrp1 protein degradation via a Dyn1-dependent pathway, revealing a previously uncovered role of Dyn1 in LPS-promoted Nrp1 protein decay. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. Effective knockdown-replace gene therapy in a novel mouse model of DNM1 developmental and epileptic encephalopathy.
- Author
-
Jones DJ, Soundararajan D, Taylor NK, Aimiuwu OV, Mathkar P, Shore A, Teoh JJ, Wang W, Sands TT, Weston MC, Harper SQ, and Frankel WN
- Subjects
- Animals, Humans, Mice, Dependovirus genetics, Disease Models, Animal, Dynamin I genetics, Dynamin I metabolism, GABAergic Neurons metabolism, Gene Knockdown Techniques, Genetic Vectors genetics, Genetic Vectors administration & dosage, Mutation, RNA Interference, RNA, Small Interfering genetics, Epilepsy therapy, Epilepsy genetics, Genetic Therapy methods
- Abstract
Effective gene therapy for gain-of-function or dominant-negative disease mutations may require eliminating expression of the mutant copy together with wild-type replacement. We evaluated such a knockdown-replace strategy in a mouse model of DNM1 disease, a debilitating and intractable neurodevelopmental epilepsy. To challenge the approach robustly, we expressed a patient-based variant in GABAergic neurons-which resulted in growth delay and lethal seizures evident by postnatal week three-and delivered to newborn pups an AAV9-based vector encoding a ubiquitously expressed, Dnm1-specific interfering RNA (RNAi) bivalently in tail-to-tail configuration with a neuron-specific, RNAi-resistant, codon-optimized Dnm1 cDNA. Pups receiving RNAi or cDNA alone fared no better than untreated pups, whereas the vast majority of mutants receiving modest doses survived with almost full growth recovery. Synaptic recordings of cortical neurons derived from treated pups revealed that significant alterations in transmission from inhibitory to excitatory neurons were rectified by bivalent vector application. To examine the mutant transcriptome and impact of treatment, we used RNA sequencing and functional annotation clustering. Mutants displayed abnormal expression of more than 1,000 genes in highly significant and relevant functional clusters, clusters that were abrogated by treatment. Together these results suggest knockdown-replace as a potentially effective strategy for treating DNM1 and related genetic neurodevelopmental disease., Competing Interests: Declaration of interests A patent application was submitted (Appl. No. 63/639,576: Products and methods to inhibit expression of dynamin-1 variants and replace dynamin-1). Inventors: S.Q.H., N.T., and W.N.F., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Quantitative Fluorescent in situ Hybridization Reveals Differential Transcription Profile Sharpening of Endocytic Proteins in Cochlear Hair Cells Upon Maturation
- Author
-
Guobin Huang and Stephanie Eckrich
- Subjects
fluorescent in situ hybridization ,cochlea ,hair cells ,endocytosis ,dynamin-1 ,dynamin-3 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The organ of Corti (OC) comprises two types of sensory cells: outer hair cells (OHCs) and inner hair cells (IHCs). While both are mechanotransducers, OHCs serve as cochlear amplifiers, whereas IHCs transform sound into transmitter release. Reliable sound encoding is ensured by indefatigable exocytosis of synaptic vesicles associated with efficient replenishment of the vesicle pool. Vesicle reformation requires retrieval of vesicle membrane from the hair cell’s membrane via endocytosis. So far, the protein machinery for endocytosis in pre-mature and terminally differentiated hair cells has only partially been deciphered. Here, we studied three endocytic proteins, dynamin-1, dynamin-3, and endophilin-A1, by assessing their transcription profiles in pre-mature and mature mouse OCs. State-of-the-art RNAscope® fluorescent in situ hybridization (FISH) of whole-mount OCs was used for quantification of target mRNAs on single-cell level. We found that pre-mature IHCs contained more mRNA transcripts of dnm1 (encoding dynamin-1) and sh3gl2 (endophilin-A1), but less of dnm3 (dynamin-3) than OHCs. These differential transcription profiles between OHCs and IHCs were sharpened upon maturation. It is noteworthy that low but heterogeneous signal numbers were found between individual negative controls, which highlights the importance of corresponding analyses in RNAscope® assays. Complementary immunolabeling revealed strong expression of dynamin-1 in the soma of mature IHCs, which was much weaker in pre-mature IHCs. By contrast, dynamin-3 was predominantly found in the soma and at the border of the cuticular plates of pre-mature and mature OHCs. In summary, using quantitative RNAscope® FISH and immunohistochemistry on whole-mount tissue of both pre-mature and mature OCs, we disclosed the cellular upregulation of endocytic proteins at the level of transcription/translation during terminal differentiation of the OC. Dynamin-1 and endophilin-A1 likely contribute to the strengthening of the endocytic machinery in IHCs after the onset of hearing, whereas expression of dynamin-3 at the cuticular plate of pre-mature and mature OHCs suggests its possible involvement in activity-independent apical endocytosis.
- Published
- 2021
- Full Text
- View/download PDF
8. Quantitative Fluorescent in situ Hybridization Reveals Differential Transcription Profile Sharpening of Endocytic Proteins in Cochlear Hair Cells Upon Maturation.
- Author
-
Huang, Guobin and Eckrich, Stephanie
- Subjects
FLUORESCENCE in situ hybridization ,ENDOCYTOSIS ,HAIR cells ,CORTI'S organ ,SYNAPTIC vesicles ,PROTEINS - Abstract
The organ of Corti (OC) comprises two types of sensory cells: outer hair cells (OHCs) and inner hair cells (IHCs). While both are mechanotransducers, OHCs serve as cochlear amplifiers, whereas IHCs transform sound into transmitter release. Reliable sound encoding is ensured by indefatigable exocytosis of synaptic vesicles associated with efficient replenishment of the vesicle pool. Vesicle reformation requires retrieval of vesicle membrane from the hair cell's membrane via endocytosis. So far, the protein machinery for endocytosis in pre-mature and terminally differentiated hair cells has only partially been deciphered. Here, we studied three endocytic proteins, dynamin-1, dynamin-3, and endophilin-A1, by assessing their transcription profiles in pre-mature and mature mouse OCs. State-of-the-art RNAscope
® fluorescent in situ hybridization (FISH) of whole-mount OCs was used for quantification of target mRNAs on single-cell level. We found that pre-mature IHCs contained more mRNA transcripts of dnm1 (encoding dynamin-1) and sh3gl2 (endophilin-A1), but less of dnm3 (dynamin-3) than OHCs. These differential transcription profiles between OHCs and IHCs were sharpened upon maturation. It is noteworthy that low but heterogeneous signal numbers were found between individual negative controls, which highlights the importance of corresponding analyses in RNAscope® assays. Complementary immunolabeling revealed strong expression of dynamin-1 in the soma of mature IHCs, which was much weaker in pre-mature IHCs. By contrast, dynamin-3 was predominantly found in the soma and at the border of the cuticular plates of pre-mature and mature OHCs. In summary, using quantitative RNAscope® FISH and immunohistochemistry on whole-mount tissue of both pre-mature and mature OCs, we disclosed the cellular upregulation of endocytic proteins at the level of transcription/translation during terminal differentiation of the OC. Dynamin-1 and endophilin-A1 likely contribute to the strengthening of the endocytic machinery in IHCs after the onset of hearing, whereas expression of dynamin-3 at the cuticular plate of pre-mature and mature OHCs suggests its possible involvement in activity-independent apical endocytosis. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
9. BTBD9 and dopaminergic dysfunction in the pathogenesis of restless legs syndrome.
- Author
-
Lyu, Shangru, Doroodchi, Atbin, Xing, Hong, Sheng, Yi, DeAndrade, Mark P., Yang, Youfeng, Johnson, Tracy L., Clemens, Stefan, Yokoi, Fumiaki, Miller, Michael A., Xiao, Rui, and Li, Yuqing
- Subjects
- *
RESTLESS legs syndrome , *PATHOLOGY , *DOPAMINE receptors , *KNOCKOUT mice , *NERVOUS system , *CATAPLEXY - Abstract
Restless legs syndrome (RLS) is characterized by an urge to move legs, usually accompanied by uncomfortable sensations. RLS symptoms generally happen at night and can be relieved by movements. Genetic studies have linked polymorphisms in BTBD9 to a higher risk of RLS. Knockout of BTBD9 homolog in mice (Btbd9) and fly results in RLS-like phenotypes. A dysfunctional dopaminergic system is associated with RLS. However, the function of BTBD9 in the dopaminergic system and RLS is not clear. Here, we made use of the simple Caenorhabditis elegans nervous system. Loss of hpo-9, the worm homolog of BTBD9, resulted in hyperactive egg-laying behavior. Analysis of genetic interactions between hpo-9 and genes for dopamine receptors (dop-1, dop-3) indicated that hpo-9 and dop-1 worked similarly. Reporter assays of dop-1 and dop-3 revealed that hpo-9 knockout led to a significant increase of DOP-3 expression. This appears to be evolutionarily conserved in mice with an increased D2 receptor (D2R) mRNA in the striatum of the Btbd9 knockout mice. Furthermore, the striatal D2R protein was significantly decreased and Dynamin I was increased. Overall, activities of DA neurons in the substantia nigra were not altered, but the peripheral D1R pathway was potentiated in the Btbd9 knockout mice. Finally, we generated and characterized the dopamine neuron-specific Btbd9 knockout mice and detected an active-phase sleepiness, suggesting that dopamine neuron-specific loss of Btbd9 is sufficient to disturb the sleep. Our results suggest that increased activities in the D1R pathway, decreased activities in the D2R pathway, or both may contribute to RLS. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
10. Clinical Assessments and EEG Analyses of Encephalopathies Associated With Dynamin-1 Mutation
- Author
-
Hua Li, Fang Fang, Manting Xu, Zhimei Liu, Ji Zhou, Xiaohui Wang, Xiaofei Wang, and Tongli Han
- Subjects
epileptic encephalopathy ,dynamin-1 ,mutation ,electroencephalogram, children ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Epileptic encephalopathy, caused by mutations in the dynamin-1 (DNM1; NM_004408) gene, is a newly identified neurologic disorder in children. Thus far, the full clinical and electroencephalographic features of children with DNM1 mutation-related epileptic encephalopathy have not been established. The aim of this study is to characterize the phenotypic, genetic, and electroencephalographic features of children with DNM1 mutation-related epileptic encephalopathy. Here, we investigated a patient with a novel pathogenic DNM1 variant, who received treatment in Beijing Children's Hospital and had detailed clinical, EEG, and genetic information. Conversely, we performed an extensive literature search in PubMed, EMBASE, Cochrane Central Register of Controlled Trials, Chinese BioMedical Literature Database, China National Knowledge Infrastructure, and Wanfang Database using the term “DNM1” and were able to find 32 cases reported in nine articles (in English) from January 2013 to December 2018. The clinical features of 33 cases with pathogenic DNM1 variants were analyzed and the results showed that patients carrying pathogenic variants in the GTPase or middle domains present with epileptic encephalopathy and severe neurodevelopmental symptoms. Patients carrying pathogenic variants in both domains exhibited comparable phenotypes.
- Published
- 2019
- Full Text
- View/download PDF
11. RNAi-Based Gene Therapy Rescues Developmental and Epileptic Encephalopathy in a Genetic Mouse Model
- Author
-
Sabrina Petri, Jia Jie Teoh, Scott Q. Harper, Nettie K. Pyne, Megha Sah, Chana Rosenthal-Weiss, Osasumwen V. Aimiuwu, Mu Yang, Allison M. Fowler, Wayne N. Frankel, and Ayla Kanber
- Subjects
dynamin-1 ,Genetic enhancement ,Lennox-Gastaut syndrome ,Bioinformatics ,Epilepsy ,Mice ,0302 clinical medicine ,RNA interference ,Drug Discovery ,Medicine ,RNA interferance ,Dynamin I ,0303 health sciences ,Dependovirus ,gene therapy ,Hypotonia ,Infusions, Intraventricular ,Treatment Outcome ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,RNA Interference ,medicine.symptom ,AAV9 ,infantile spasms ,Ataxia ,adeno-associated virus 9 ,Genetic Vectors ,Viral vector ,Gene product ,03 medical and health sciences ,Genetics ,Animals ,Humans ,developmental and epileptic encephalopathy ,Molecular Biology ,030304 developmental biology ,DEE ,Pharmacology ,business.industry ,DNM1 ,Genetic Therapy ,medicine.disease ,Disease Models, Animal ,MicroRNAs ,Animals, Newborn ,epilepsy ,business ,Epileptic Syndromes ,Lennox–Gastaut syndrome - Abstract
Developmental and epileptic encephalopathy (DEE) associated with de novo variants in the gene encoding dynamin-1 (DNM1) is a severe debilitating disease with no pharmacological remedy. Like most genetic DEEs, the majority of DNM1 patients suffer from therapy-resistant seizures and comorbidities such as intellectual disability, developmental delay, and hypotonia. We tested RNAi gene therapy in the Dnm1 fitful mouse model of DEE using a Dnm1-targeted therapeutic microRNA delivered by a self-complementary adeno-associated virus vector. Untreated or control-injected fitful mice have growth delay, severe ataxia, and lethal tonic-clonic seizures by 3 weeks of age. These major impairments are mitigated following a single treatment in newborn mice, along with key underlying cellular features including gliosis, cell death, and aberrant neuronal metabolic activity typically associated with recurrent seizures. Our results underscore the potential for RNAi gene therapy to treat DNM1 disease and other genetic DEEs where treatment would require inhibition of the pathogenic gene product., Graphical Abstract, Toxic mutations in DNM1, a gene involved in the proper functioning of brain cells, cause developmental and epileptic encephalopathy—a severe class of epilepsy with no cure. Aimiuwu and colleagues show that a single gene therapy treatment via RNA interference eliminated the toxic allele sufficiently to curb severe lethal seizures, eliminate motor and developmental deficits, and extend survival of treated mice.
- Published
- 2020
12. Lack of evidence in neurite growth in the gerbil hippocampal CA1 region 15 days after transient forebrain ischemia.
- Author
-
Yoo, Dae Young, Kwon, Hyun Jung, Lee, Kwon Young, Jung, Hyo Young, Kim, Jong Whi, Park, Joon Ha, Choi, Jung Hoon, Yoon, Yeo Sung, Won, Moo-Ho, Kim, Dae Won, and Hwang, In Koo
- Subjects
- *
CEREBRAL ischemia , *NEURON development , *GERBILS , *HIPPOCAMPUS (Brain) , *GLUCOSE transporters , *DYNAMIN (Genetics) , *ANIMAL models of ischemia , *SURGERY - Abstract
Transient forebrain ischemia promotes a robust increase in neuroblast differentiation in the hippocampal dentate gyrus that peaks 7–15 days after the surgery. In this study, we compared the glucose transporter 3 (GLUT3)-dependent glucose utilization and the dynamin-1 (DNM1)-dependent neurite growth in the hippocampus of Mongolian gerbils 15 days after the induction of transient forebrain ischemia. The animals were subjected to a 5 min transient ischemia protocol and sacrificed 15 days after the surgery. Both doublecortin (DCX) immunoreactive neuroblasts and DCX total protein levels were abundantly increased in the ischemic group compared to the levels observed in the control group. In addition, animals in the ischemic group showed elevated GLUT3 immunoreactivity in the subgranular zone of the dentate gyrus compared to animals in the control group. Based on the double immunofluorescent study, increased DCX-immunoreactive neuroblasts were co-localized with GLUT3-immunoreactive components in the dentate gyrus. However, both the immunoreactivity and the total protein levels of DNM1 were significantly decreased in the dentate gyrus and hippocampal CA1 regions of the ischemic group. These results suggest that the regeneration process such as neurite growth is lacking in the hippocampus 15 days after ischemia/reperfusion although neuroblasts production and glucose utilization increased in the hippocampus. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
13. Antibody Characterization Report for Dynamin-1
- Author
-
Ayoubi, Riham, Alshafie, Walaa, You, Zhipeng, Durcan, Thomas M., McPherson, Peter S., and Laflamme, Carl
- Subjects
Dynamin-1 ,antibody characterization ,Q05193 ,DNM1 ,antibody validation ,3. Good health - Abstract
Head to head comparison of available commercial antibodies against Dynamin-1 by immunoblot (Western blot), immunoprecipitation and immunofluorescence.
- Published
- 2021
- Full Text
- View/download PDF
14. Quantitative Fluorescent in situ Hybridization Reveals Differential Transcription Profile Sharpening of Endocytic Proteins in Cochlear Hair Cells Upon Maturation
- Author
-
Stephanie Eckrich and Guobin Huang
- Subjects
0301 basic medicine ,hair cells ,dynamin-1 ,Endocytic cycle ,cochlea ,dynamin-3 ,Cuticular plate ,Endocytosis ,Synaptic vesicle ,Exocytosis ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,endocytosis ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Cochlea ,Original Research ,Chemistry ,terminal differentiation ,Cell biology ,endophilin-A1 ,030104 developmental biology ,medicine.anatomical_structure ,Organ of Corti ,fluorescent in situ hybridization ,Hair cell ,sense organs ,030217 neurology & neurosurgery ,Neuroscience - Abstract
The organ of Corti (OC) comprises two types of sensory cells: outer hair cells (OHCs) and inner hair cells (IHCs). While both are mechanotransducers, OHCs serve as cochlear amplifiers, whereas IHCs transform sound into transmitter release. Reliable sound encoding is ensured by indefatigable exocytosis of synaptic vesicles associated with efficient replenishment of the vesicle pool. Vesicle reformation requires retrieval of vesicle membrane from the hair cell’s membrane via endocytosis. So far, the protein machinery for endocytosis in pre-mature and terminally differentiated hair cells has only partially been deciphered. Here, we studied three endocytic proteins, dynamin-1, dynamin-3, and endophilin-A1, by assessing their transcription profiles in pre-mature and mature mouse OCs. State-of-the-art RNAscope® fluorescent in situ hybridization (FISH) of whole-mount OCs was used for quantification of target mRNAs on single-cell level. We found that pre-mature IHCs contained more mRNA transcripts of dnm1 (encoding dynamin-1) and sh3gl2 (endophilin-A1), but less of dnm3 (dynamin-3) than OHCs. These differential transcription profiles between OHCs and IHCs were sharpened upon maturation. It is noteworthy that low but heterogeneous signal numbers were found between individual negative controls, which highlights the importance of corresponding analyses in RNAscope® assays. Complementary immunolabeling revealed strong expression of dynamin-1 in the soma of mature IHCs, which was much weaker in pre-mature IHCs. By contrast, dynamin-3 was predominantly found in the soma and at the border of the cuticular plates of pre-mature and mature OHCs. In summary, using quantitative RNAscope® FISH and immunohistochemistry on whole-mount tissue of both pre-mature and mature OCs, we disclosed the cellular upregulation of endocytic proteins at the level of transcription/translation during terminal differentiation of the OC. Dynamin-1 and endophilin-A1 likely contribute to the strengthening of the endocytic machinery in IHCs after the onset of hearing, whereas expression of dynamin-3 at the cuticular plate of pre-mature and mature OHCs suggests its possible involvement in activity-independent apical endocytosis.
- Published
- 2021
- Full Text
- View/download PDF
15. Screening and identification of dynamin-1 interacting proteins in rat brain synaptosomes.
- Author
-
Zhang, Ciliu, Omran, Ahmed Galal, He, Fang, Deng, Xiaolu, Wu, Lei, Peng, Jing, and Yin, Fei
- Subjects
- *
GUANOSINE triphosphatase , *DYNAMIN (Genetics) , *SYNAPTOSOMES , *GENE expression , *PROTEIN-protein interactions , *SYNAPTIC vesicles , *LABORATORY rats - Abstract
Abstract: Dynamin-1 is a multi-domain GTPase that is crucial for the fission stage of synaptic vesicle recycling and vesicle trafficking. In this study, we constructed prokaryotic expression plasmids for the four functional domains of dynamin-1, which are pGEX-4T-2-PH, pGEX-4T-2-PRD, pGEX-4T-2-GED and pGEX-4T-2-GTPase. Glutathione S-transferase pull-down, co-immunoprecipitation (co-IP), and liquid chromatography/mass spectrometry were used to screen and identify dynamin-1 interacting proteins in rat brain synaptosomes. We identified a set of 63 candidate protein interactions, including 36 proteins interacting with dynamin-1 C-terminal proline-rich domain (PRD), 14 with pleckstrin-homology domain (PH), 7 with GTPase effector domain (GED) and 6 with GTPase domain, consisting of synaptic vesicle-associated proteins, cytoskeletal proteins, metabolic enzymes and other proteins. We selected three previously unreported dynamin-1 interacting proteins to verify their interaction with dynamin-1 under native conditions. Using co-IP, we found that Rab GDP-dissociation inhibitor (Rab GDI) and chloride channel 3 (ClC-3) do interact with dynamin-1, but not with TUC-4b (the TOAD-64/Ulip/CRMP (TUC) family member). Those novel interactions detected in our study offer valuable insight into the protein–protein interacting network that could enhance our understanding of dynamin-1 mediated synaptic vesicle recycling. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
16. Antibody induction of lupus-like neuropsychiatric manifestations
- Author
-
Lawrence, David A., Bolivar, Valerie J., Hudson, Chad A., Mondal, Tapan K., and Pabello, Nina G.
- Subjects
- *
AUTOIMMUNE diseases , *SYSTEMIC lupus erythematosus , *ANTIGENS , *AUTOANTIBODIES - Abstract
Abstract: Although systemic lupus erythematosus (SLE) is usually evaluated with regard to autoimmune reactivity toward the kidney, there are multiple psychiatric abnormalities associated with this autoimmune disease. Lupus-prone male NZM88 mice, derived from NZB/NZW F1 mice, develop early neuropsychiatric manifestations without any signs of nephritis. In addition to the usual repertoire of antibody specificities, including autoantibodies to dsDNA and renal antigens, mice of this inbred strain express autoantibodies to numerous brain antigens. Here, we show that autoantibodies to brain antigens, assessed by Western analysis, are as individually varied as are the diverse neuropsychiatric manifestations observed in SLE patients. Additionally, a monoclonal antibody derived from the spleen of an untreated NZM88 male when injected into healthy BALB/cByJ, but not C57BL/6J, mice induced behaviors similar to those of lupus-prone NZM88 mice. This monoclonal antibody, which is specific to dynamin-1, binds preferentially in BALB/cByJ cortex and induces substantial expression of cytokines mainly in the hypothalamus. Thus, an antibody to just one brain antigen can induce multiple behavioral changes, and multiple autoantibodies to different brain antigens exist in lupus-prone mice; however, susceptibility to the induction of neurobehavioral deficits is dependent on host genetics. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
17. Receptor-type PTP-NP inhibition of Dynamin-1 GTPase activity is associated with neuronal depolarization
- Author
-
Jiang, Shuxian, Avraham, Hava Karsenty, Kim, Tae-Aug, Rogers, Rick A., and Avraham, Shalom
- Subjects
- *
GREEN fluorescent protein , *ENDOCYTOSIS , *TYROSINE , *PROTEIN kinases - Abstract
Abstract: Dynamin-1 is a GTP-hydrolyzing protein and a key element in the clathrin-mediated endocytosis of secretory granules and neurovesicles at the plasma membrane. The unique receptor-like protein tyrosine phosphatase, PTP-NP/Phogrin/IAR/IA-2, is associated with neuroendocrine secretory granules and is highly expressed in the brain. Here, we show by confocal microscopy and biochemical studies that PTP-NP rapidly associates with Dynamin-1 in a depolarization-dependent manner and regulates Dynamin-1 GTPase activity upon KCl depolarization of rat primary hippocampal neurons. Depolarization of primary neurons induced direct association of PTP-NP with Dynamin-1 within 30 s. This association resulted in significant inhibition of Dynamin-1 GTPase activity (∼75% inhibition). Mutation within the phosphatase domain of PTP-NP (PTP-NPD947A) abolished the direct interaction of PTP-NP with Dynamin-1 and failed to inhibit Dynamin-1 GTPase activity. To further confirm the endogenous interaction of Dynamin-1 with wild-type PTP-NP, Dynamin-1 was purified biochemically from rat brain and its interaction with purified PTP-NP was analyzed. Highly purified Dynamin-1 specifically associated with wild-type PTP-NP and not with mutated PTP-NP, resulting in significant inhibition (∼70%) of Dynamin-1 GTPase activity. This is the first report to suggest a novel function of this unique receptor-type tyrosine phosphatase as a potential regulator of Dynamin-1 GTPase activity upon neuronal depolarization. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
18. Phospholipase C-γ1 is a guanine nucleotide exchange factor for dynamin-1 and enhances dynamin-1-dependent epidermal growth factor receptor endocytosis.
- Author
-
Jang Hyun Choi, Jong Bae Park, Sun Sik Bae, Sanguk Yun, Hyeon Soo Kim, Won-Pyo Hong, Il-Shin Kim, Jae Ho Kim, Mi Young Han, Sung Ho Ryu, Patterson, Randen L., Snyder, Solomon H., and Suh, Pann-Ghill
- Subjects
- *
PHOSPHOLIPASES , *MOLECULES , *HOMOLOGY (Biology) , *CELLS , *ENDOCYTOSIS , *G proteins - Abstract
Phospholipase C-γ1 (PLC-γ1), which interacts with a variety of signaling molecules through its two Src homology (SH) 2 domains and a single SH3 domain has been implicated in the regulation of many cellular functions. We demonstrate that PLC-γ1 acts as a guanine nucleotide exchange factor (GEF) of dynamin-1, a 100 kDa GTPase protein, which is involved in clathrin-mediated endocytosis of epidermal growth factor (EGF) receptor. Overexpression of PLC-γ1 increases endocytosis of the EGF receptor by increasing guanine nucleotide exchange activity of dynamin-1. The GEF activity of PLC-γ1 is mediated by the direct interaction of its SH3 domain with dynamin-1. EGF- dependent activation of ERK and serum response element (SRE) are both up-regulated in PC12 cells stably overexpressing PLC-γ1, but knockdown of PLC-γ1 by siRNA significantly reduces ERK activation. These results establish a new role for PLC-γ1 in the regulation of endocytosis and suggest that endocytosis of activated EGF receptors may mediate PLC-γl-dependent proliferation. [ABSTRACT FROM AUTHOR]
- Published
- 2004
- Full Text
- View/download PDF
19. Clinical Assessments and EEG Analyses of Encephalopathies Associated With Dynamin-1 Mutation
- Author
-
Tongli Han, Xiaofei Wang, Hua Li, Manting Xu, Zhimei Liu, Xiaohui Wang, Fang Fang, and Ji Zhou
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,dynamin-1 ,Electroencephalography ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Pharmacology (medical) ,Dynamin ,Original Research ,Pharmacology ,Mutation ,medicine.diagnostic_test ,business.industry ,Epileptic encephalopathy ,lcsh:RM1-950 ,electroencephalogram, children ,Knowledge infrastructure ,3. Good health ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,epileptic encephalopathy ,030220 oncology & carcinogenesis ,mutation ,business - Abstract
Epileptic encephalopathy, caused by mutations in the dynamin-1 (DNM1; NM_004408) gene, is a newly identified neurologic disorder in children. Thus far, the full clinical and electroencephalographic features of children with DNM1 mutation-related epileptic encephalopathy have not been established. The aim of this study is to characterize the phenotypic, genetic, and electroencephalographic features of children with DNM1 mutation-related epileptic encephalopathy. Here, we investigated a patient with a novel pathogenic DNM1 variant, who received treatment in Beijing Children's Hospital and had detailed clinical, EEG, and genetic information. Conversely, we performed an extensive literature search in PubMed, EMBASE, Cochrane Central Register of Controlled Trials, Chinese BioMedical Literature Database, China National Knowledge Infrastructure, and Wanfang Database using the term “DNM1” and were able to find 32 cases reported in nine articles (in English) from January 2013 to December 2018. The clinical features of 33 cases with pathogenic DNM1 variants were analyzed and the results showed that patients carrying pathogenic variants in the GTPase or middle domains present with epileptic encephalopathy and severe neurodevelopmental symptoms. Patients carrying pathogenic variants in both domains exhibited comparable phenotypes.
- Published
- 2019
20. De Novo Mutations in Synaptic Transmission Genes Including DNM1 Cause Epileptic Encephalopathies
- Author
-
Silke Appenzeller, Rudi Balling, Nina Barisic, Stéphanie Baulac, Hande Caglayan, Dana Craiu, Peter De Jonghe, Christel Depienne, Petia Dimova, Tania Djémié, Padhraig Gormley, Renzo Guerrini, Ingo Helbig, Helle Hjalgrim, Dorota Hoffman-Zacharska, Johanna Jähn, Karl Martin Klein, Bobby Koeleman, Vladimir Komarek, Roland Krause, Gregor Kuhlenbäumer, Eric Leguern, Anna-Elina Lehesjoki, Johannes R. Lemke, Holger Lerche, Tarja Linnankivi, Carla Marini, Patrick May, Rikke S. Møller, Hiltrud Muhle, Deb Pal, Aarno Palotie, Manuela Pendziwiat, Angela Robbiano, Filip Roelens, Felix Rosenow, Kaja Selmer, Jose M. Serratosa, Sanjay Sisodiya, Ulrich Stephani, Katalin Sterbova, Pasquale Striano, Arvid Suls, Tiina Talvik, Sarah von Spiczak, Yvonne Weber, Sarah Weckhuysen, Federico Zara, Bassel Abou-Khalil, Brian K. Alldredge, Eva Andermann, Frederick Andermann, Dina Amrom, Jocelyn F. Bautista, Samuel F. Berkovic, Judith Bluvstein, Alex Boro, Gregory Cascino, Damian Consalvo, Patricia Crumrine, Orrin Devinsky, Dennis Dlugos, Michael P. Epstein, Miguel Fiol, Nathan B. Fountain, Jacqueline French, Daniel Friedman, Eric B. Geller, Tracy Glauser, Simon Glynn, Kevin Haas, Sheryl R. Haut, Jean Hayward, Sandra L. Helmers, Sucheta Joshi, Andres Kanner, Heidi E. Kirsch, Robert C. Knowlton, Eric H. Kossoff, Rachel Kuperman, Ruben Kuzniecky, Daniel H. Lowenstein, Shannon M. McGuire, Paul V. Motika, Edward J. Novotny, Ruth Ottman, Juliann M. Paolicchi, Jack Parent, Kristen Park, Annapurna Poduri, Lynette Sadleir, Ingrid E. Scheffer, Renée A. Shellhaas, Elliott Sherr, Jerry J. Shih, Rani Singh, Joseph Sirven, Michael C. Smith, Joe Sullivan, Liu Lin Thio, Anu Venkat, Eileen P.G. Vining, Gretchen K. Von Allmen, Judith L. Weisenberg, Peter Widdess-Walsh, Melodie R. Winawer, Andrew S. Allen, Patrick Cossette, Norman Delanty, Evan E. Eichler, David B. Goldstein, Yujun Han, Erin L. Heinzen, Michael R. Johnson, Anthony G. Marson, Heather C. Mefford, Sahar Esmaeeli Nieh, Terence J. O’Brien, Stephen Petrou, Slavé Petrovski, Elizabeth K. Ruzzo, Neuroscience Center, Research Programs Unit, Department of Medical and Clinical Genetics, Research Programme for Molecular Neurology, Institute for Molecular Medicine Finland, Genomics of Neurological and Neuropsychiatric Disorders, EuroEPINOMICS-RES Consortium, Epilepsy Phenome Genome Project, and Epi4K Consortium
- Subjects
0301 basic medicine ,Male ,Proband ,INTELLECTUAL DISABILITY ,Type I ,Bioinformatics ,medicine.disease_cause ,Infantile ,Synaptic Transmission ,Spasms ,Cohort Studies ,Epilepsy ,0302 clinical medicine ,Receptors ,SCHIZOPHRENIA ,STXBP1 ,Exome ,Gene Regulatory Networks ,Protein Interaction Maps ,Dynamin I ,Genetics (clinical) ,Genetics ,0303 health sciences ,Mutation ,education.field_of_study ,Medicine (all) ,Genome project ,Fatty Acid Synthase, Type I ,Fatty Acid Synthase ,Female ,APHASIA ,Spasms, Infantile ,DYNAMIN-1 ,EPILEPSIES ,ENDOCYTOSIS ,Population ,SPECTRUM DISORDERS ,Phenome ,Neurotransmission ,Biology ,GNAO1 ,Article ,03 medical and health sciences ,GRIN2A MUTATIONS ,medicine ,Humans ,AUTISM ,Infant, Newborn ,Lennox Gastaut Syndrome ,Receptors, GABA-B ,Ryanodine Receptor Calcium Release Channel ,education ,Gene ,De novo mutations ,030304 developmental biology ,GABA-B ,3112 Neurosciences ,Infant ,Correction ,Newborn ,medicine.disease ,Human genetics ,030104 developmental biology ,DNM1 ,PATTERNS ,Human medicine ,030217 neurology & neurosurgery ,Lennox–Gastaut syndrome - Abstract
Emerging evidence indicates that epileptic encephalopathies are genetically highly heterogeneous, underscoring the need for large cohorts of well-characterized individuals to further define the genetic landscape. Through a collaboration between two consortia (EuroEPINOMICS and Epi4K/EPGP), we analyzed exome-sequencing data of 356 trios with the "classical" epileptic encephalopathies, infantile spasms and Lennox Gastaut syndrome, including 264 trios previously analyzed by the Epi4K/EPGP consortium. In this expanded cohort, we find 429 de nova mutations, including de novo mutations in DNM1 in five individuals and de nova mutations in GABBR2, FASN, and RYR3 in two individuals each. Unlike previous studies, this cohort is sufficiently large to show a significant excess of de nova mutations in epileptic encephalopathy probands compared to the general population using a likelihood analysis (p = 8.2 x 10(-4)), supporting a prominent role for de novo mutations in epileptic encephalopathies. We bring statistical evidence that mutations in DNM1 cause epileptic encephalopathy, find suggestive evidence for a role of three additional genes, and show that at least 12% of analyzed individuals have an identifiable causal de novo mutation. Strikingly, 75% of mutations in these probands are predicted to disrupt a protein involved in regulating synaptic transmission, and there is a significant enrichment of de nova mutations in genes in this pathway in the entire cohort as well. These findings emphasize an important role for synaptic dysregulation in epileptic encephalopathies, above and beyond that caused by ion channel dysfunction.
- Published
- 2017
- Full Text
- View/download PDF
21. RNAi-Based Gene Therapy Rescues Developmental and Epileptic Encephalopathy in a Genetic Mouse Model.
- Author
-
Aimiuwu OV, Fowler AM, Sah M, Teoh JJ, Kanber A, Pyne NK, Petri S, Rosenthal-Weiss C, Yang M, Harper SQ, and Frankel WN
- Subjects
- Animals, Animals, Newborn, Dependovirus genetics, Disease Models, Animal, Epileptic Syndromes genetics, Epileptic Syndromes pathology, Genetic Vectors administration & dosage, Humans, Infusions, Intraventricular, Mice, MicroRNAs administration & dosage, RNA Interference, Treatment Outcome, Dynamin I genetics, Epileptic Syndromes therapy, Genetic Therapy methods, MicroRNAs genetics
- Abstract
Developmental and epileptic encephalopathy (DEE) associated with de novo variants in the gene encoding dynamin-1 (DNM1) is a severe debilitating disease with no pharmacological remedy. Like most genetic DEEs, the majority of DNM1 patients suffer from therapy-resistant seizures and comorbidities such as intellectual disability, developmental delay, and hypotonia. We tested RNAi gene therapy in the Dnm1 fitful mouse model of DEE using a Dnm1-targeted therapeutic microRNA delivered by a self-complementary adeno-associated virus vector. Untreated or control-injected fitful mice have growth delay, severe ataxia, and lethal tonic-clonic seizures by 3 weeks of age. These major impairments are mitigated following a single treatment in newborn mice, along with key underlying cellular features including gliosis, cell death, and aberrant neuronal metabolic activity typically associated with recurrent seizures. Our results underscore the potential for RNAi gene therapy to treat DNM1 disease and other genetic DEEs where treatment would require inhibition of the pathogenic gene product., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. Life-Long Effects of Perinatal Asphyxia on Stress-Induced Proteins and Dynamin 1 in Rat Brain
- Author
-
Kitzmueller, Erwin, Krapfenbauer, Kurt, Hoeger, Harald, Weitzdoerfer, Rachel, Lubec, Gert, and Lubec, Barbara
- Published
- 2004
- Full Text
- View/download PDF
23. Crosstalk between CLCb/Dyn1-Mediated Adaptive Clathrin-Mediated Endocytosis and Epidermal Growth Factor Receptor Signaling Increases Metastasis.
- Author
-
Chen, Ping-Hung, Bendris, Nawal, Hsiao, Yi-Jing, Reis, Carlos R., Mettlen, Marcel, Chen, Hsuan-Yu, Yu, Sung-Liang, and Schmid, Sandra L.
- Subjects
- *
CLATHRIN , *EPIDERMAL growth factor receptors , *METASTASIS , *CROSSTALK , *CELLULAR signal transduction , *PROGNOSIS - Abstract
Summary Signaling receptors are internalized and regulated by clathrin-mediated endocytosis (CME). Two clathrin light chain isoforms, CLCa and CLCb, are integral components of the endocytic machinery whose differential functions remain unknown. We report that CLCb is specifically upregulated in non-small-cell lung cancer (NSCLC) cells and is associated with poor patient prognosis. Engineered single CLCb-expressing NSCLC cells, as well as “switched” cells that predominantly express CLCb, exhibit increased rates of CME and altered clathrin-coated pit dynamics. This “adaptive CME” resulted from upregulation of dynamin-1 (Dyn1) and its activation through a positive feedback loop involving enhanced epidermal growth factor (EGF)-dependent Akt/GSK3β phosphorylation. CLCb/Dyn1-dependent adaptive CME selectively altered EGF receptor trafficking, enhanced cell migration in vitro, and increased the metastatic efficiency of NSCLC cells in vivo. We define molecular mechanisms for adaptive CME in cancer cells and a role for the reciprocal crosstalk between signaling and CME in cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
24. Antibody Characterization Report for Dynamin-1
- Author
-
Ayoubi, Riham, Alshafie, Walaa, You, Zhipeng, Durcan, Thomas M., McPherson, Peter S., and Laflamme, Carl
- Subjects
Dynamin-1 ,antibody characterization ,Q05193 ,DNM1 ,antibody validation ,3. Good health - Abstract
Head to head comparison of available commercial antibodies against Dynamin-1 by immunoblot (Western blot), immunoprecipitation and immunofluorescence.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.