1
|
Witke W, Di Domenico M, Maggi L, Di Nardo A, Stein V, Pilo Boyl P. Autism spectrum disorder related phenotypes in a mouse model lacking the neuronal actin binding protein profilin 2. Front Cell Neurosci 2025; 19:1540989. [PMID: 40078324 PMCID: PMC11897305 DOI: 10.3389/fncel.2025.1540989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Profilin 2 (PFN2) is an actin binding protein highly expressed in the brain that participates in actin dynamics. It has been shown in vitro and in vivo that in neurons it functions both post-synaptically to shape and maintain dendritic arborizations and spine density and plasticity, as well as pre-synaptically to regulate vesicle exocytosis. PFN2 was also found in protein complexes with proteins that have been implicated in or are causative of autism spectrum disorder. Methods We employ a genetically engineered knock-out mouse line for Pfn2 that we previously generated to study the mouse social, vocal and motor behavior in comparison to wild type control littermates. We also study neuronal physiology in the knock-out mouse model by means of cellular and field electrophysiological recordings in cerebellar Purkinje cells and in the Schaffer collaterals. Lastly, we study anatomical features of the cerebellum using immunofluorescence stainings. Results We show that PFN2 deficiency reproduces a number of autistic-like phenotypes in the mouse, such as social behavior impairment, stereotypic behavior, altered vocal communication, and deficits in motor performance and coordination. Our studies correlate the behavioral phenotypes with increased excitation/inhibition ratio in the brain, due to brain-wide hyperactivity of glutamatergic neurons and increased glutamate release not compensated by enhanced GABAergic neurotransmission. Consequently, lack of PFN2 caused seizures behavior and age-dependent loss of cerebellar Purkinje cells, comorbidities observed in a subset of autistic patients, which can be attributed to the effect of excessive glutamatergic neurotransmission. Discussion Our data directly link altered pre-synaptic actin dynamics to autism spectrum disorder in the mouse model and support the hypothesis that synaptic dysfunctions that asymmetrically increase the excitatory drive in neuronal circuits can lead to autistic-like phenotypes. Our findings inspire to consider novel potential pathways for therapeutic approaches in ASD.
Collapse
Affiliation(s)
- Walter Witke
- Institute of Genetics, University of Bonn, Bonn, Germany
| | | | - Laura Maggi
- Dipartimento di Fisiologia e Farmacologia, Research Center of Neuroscience “CRiN-Daniel Bovet”, University Sapienza Rome, Rome, Italy
| | | | - Valentin Stein
- Institute of Physiology II, Faculty of Medicine, University of Bonn, Bonn, Germany
| | | |
Collapse
|
2
|
Shi T, Zhou Z, Xiang T, Suo Y, Shi X, Li Y, Zhang P, Dai J, Sheng L. Cytoskeleton dysfunction of motor neuron in spinal muscular atrophy. J Neurol 2024; 272:19. [PMID: 39666039 PMCID: PMC11638312 DOI: 10.1007/s00415-024-12724-3] [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: 09/12/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deletions or mutations of survival of motor neuron 1 (SMN1) gene. To date, the mechanism of selective cell death of motor neurons as a hallmark of SMA is still unclear. The severity of SMA is dependent on the amount of survival motor neuron (SMN) protein, which is an essential and ubiquitously expressed protein involved in various cellular processes including regulation of cytoskeletal dynamics. In this review, we discuss the effect of SMN ablation on cytoskeleton organization including actin dynamics, growth cone formation, axonal stability, neurite outgrowth, microtubule stability, synaptic vesicle dynamics and neurofilament protein release in SMA. We also summarized a list of critical proteins such as profilin-2 (PFN2), plastin-3 (PLS3), stathmin-1 (STMN1), microtubule-associated protein 1B (MAP1B) and neurofilament which play an important role in modulating cytoskeleton in SMA. Our aim is to highlight how cytoskeletal defects contribute to motor neuron degeneration in SMA disease progression and concentrating on cytoskeleton dynamics may be a promising approach to develop new therapy or biomarker.
Collapse
Affiliation(s)
- Tianyu Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Zijie Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Taiyang Xiang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Yinxuan Suo
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Xiaoyan Shi
- Department of Neurology, Children's Hospital of Soochow University, Suzhou, 215025, Jiangsu, China
| | - Yaoyao Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Peng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China
| | - Jun Dai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China.
| | - Lei Sheng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang Road, Suzhou, 215004, Jiangsu, China.
| |
Collapse
|
3
|
Li J, Zhou L, Jiang Y, Gao H, Maierhaba T, Gong H. Long noncoding RNA RMRP ameliorates doxorubicin-induced apoptosis by interacting with PFN1 in a P53-Dependent manner. Mol Cell Probes 2023; 72:101937. [PMID: 37820747 DOI: 10.1016/j.mcp.2023.101937] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/27/2023] [Accepted: 10/06/2023] [Indexed: 10/13/2023]
Abstract
Doxorubicin (DOX) often causes acute or chronic cardiotoxicity during its application. LncRNA RMRP has been reported to be associated with several biological processes, such as cartilage-hair hypoplasia, but the relationship between RMRP and DOX-induced cardiotoxicity and chronic heart failure remains obscure. To test this hypothesis, GSE124401 and GSE149870 were processed for bioinformatics, and differentially expressed RMRP was then verified in the peripheral blood of 21 patients with heart failure compared with 7 controls. For in vitro validation, we used AC16 and HEK-293T cells. qPCR was used to detect the mRNA expression levels. The degree of apoptosis was detected by Western blot and TUNEL staining. Furthermore, the interaction between RMRP and PFN1 mRNA was verified by dual-luciferase reporter assays. In bioinformatics, RMRP showed significant downregulation, which was verified in clinical samples (p < 0.001) and DOX-treated AC16 models (p < 0.0001). Next, overexpression of RMRP could significantly alleviate DOX-induced apoptosis, and a potential downstream molecule of RMRP, PFN1, was also negatively associated with this change. RESCUE experiments further confirmed that PFN1 could be regulated by RMRP at both the RNA and protein levels, serving as a downstream mediator of RMRP's cardioprotective effects. This interaction was then confirmed to be a direct combination (p < 0.0001). Finally, we found that overexpression of RMRP could inhibit the expression of p53 and its phosphorylation level by suppressing PFN1. In summary, RMRP could exert cardioprotective effects via the PFN1/p53 axis, holding great promise for serving as a therapeutic target and potential biomarker.
Collapse
Affiliation(s)
- Juexing Li
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, 201508, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei Zhou
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, 201508, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuanliang Jiang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hailan Gao
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, 201508, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tuersuntuoheti Maierhaba
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, 201508, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hui Gong
- Department of Cardiology, Jinshan Hospital of Fudan University, Shanghai, 201508, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Brabec JL, Lara MK, Tyler AL, Mahoney JM. System-Level Analysis of Alzheimer's Disease Prioritizes Candidate Genes for Neurodegeneration. Front Genet 2021; 12:625246. [PMID: 33889174 PMCID: PMC8056044 DOI: 10.3389/fgene.2021.625246] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder. Since the advent of the genome-wide association study (GWAS) we have come to understand much about the genes involved in AD heritability and pathophysiology. Large case-control meta-GWAS studies have increased our ability to prioritize weaker effect alleles, while the recent development of network-based functional prediction has provided a mechanism by which we can use machine learning to reprioritize GWAS hits in the functional context of relevant brain tissues like the hippocampus and amygdala. In parallel with these developments, groups like the Alzheimer’s Disease Neuroimaging Initiative (ADNI) have compiled rich compendia of AD patient data including genotype and biomarker information, including derived volume measures for relevant structures like the hippocampus and the amygdala. In this study we wanted to identify genes involved in AD-related atrophy of these two structures, which are often critically impaired over the course of the disease. To do this we developed a combined score prioritization method which uses the cumulative distribution function of a gene’s functional and positional score, to prioritize top genes that not only segregate with disease status, but also with hippocampal and amygdalar atrophy. Our method identified a mix of genes that had previously been identified in AD GWAS including APOE, TOMM40, and NECTIN2(PVRL2) and several others that have not been identified in AD genetic studies, but play integral roles in AD-effected functional pathways including IQSEC1, PFN1, and PAK2. Our findings support the viability of our novel combined score as a method for prioritizing region- and even cell-specific AD risk genes.
Collapse
Affiliation(s)
- Jeffrey L Brabec
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Montana Kay Lara
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States
| | - Anna L Tyler
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - J Matthew Mahoney
- Department of Neurological Sciences, University of Vermont, Burlington, VT, United States.,The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
5
|
Juneja M, Azmi A, Baets J, Roos A, Jennings MJ, Saveri P, Pisciotta C, Bernard-Marissal N, Schneider BL, Verfaillie C, Chrast R, Seeman P, Hahn AF, de Jonghe P, Maudsley S, Horvath R, Pareyson D, Timmerman V. PFN2 and GAMT as common molecular determinants of axonal Charcot-Marie-Tooth disease. J Neurol Neurosurg Psychiatry 2018; 89:870-878. [PMID: 29449460 DOI: 10.1136/jnnp-2017-317562] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/24/2018] [Accepted: 01/30/2018] [Indexed: 11/04/2022]
Abstract
BACKGROUND Charcot-Marie-Tooth type 2 (CMT2) neuropathy is characterised by a vast clinical and genetic heterogeneity complicating its diagnosis and therapeutic intervention. Identification of molecular signatures that are common to multiple CMT2 subtypes can aid in developing therapeutic strategies and measuring disease outcomes. METHODS A proteomics-based approach was performed on lymphoblasts from CMT2 patients genetically diagnosed with different gene mutations to identify differentially regulated proteins. The candidate proteins were validated through real-time quantitative PCR and western blotting on lymphoblast samples of patients and controls, motor neurons differentiated from patient-derived induced pluripotent stem cells (iPSCs) and sciatic nerves of CMT2 mouse models. RESULTS Proteomic profiling of patient lymphoblasts resulted in the identification of profilin 2 (PFN2) and guanidinoacetate methyltransferase (GAMT) as commonly downregulated proteins in different genotypes compared with healthy controls. This decrease was also observed at the transcriptional level on screening 43 CMT2 patients and 22 controls, respectively. A progressive decrease in PFN2 expression with age was observed in patients, while in healthy controls its expression increased with age. Reduced PFN2 expression was also observed in motor neurons differentiated from CMT2 patient-derived iPSCs and sciatic nerves of CMT2 mice when compared with controls. However, no change in GAMT levels was observed in motor neurons and CMT2 mouse-derived sciatic nerves. CONCLUSIONS We unveil PFN2 and GAMT as molecular determinants of CMT2 with possible indications of the role of PFN2 in the pathogenesis and disease progression. This is the first study describing biomarkers that can boost the development of therapeutic strategies targeting a wider spectrum of CMT2 patients.
Collapse
Affiliation(s)
- Manisha Juneja
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium.,Institute Born Bunge, Antwerp, Belgium
| | - Abdelkrim Azmi
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Jonathan Baets
- Institute Born Bunge, Antwerp, Belgium.,VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium.,Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerpen, Belgium
| | - Andreas Roos
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK.,Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Matthew J Jennings
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Paola Saveri
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Chiara Pisciotta
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Nathalie Bernard-Marissal
- Aix Marseille University, INSERM, MMG, U1251, Marseille, France.,Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Roman Chrast
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Pavel Seeman
- DNA Laboratory, Department of Child Neurology, 2nd Medical School, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Angelika F Hahn
- Department of Clinical Neurological Sciences Centre, University Hospital, Western University, London, Ontario, Canada
| | - Peter de Jonghe
- Institute Born Bunge, Antwerp, Belgium.,VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
| | - Rita Horvath
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, C. Besta Neurological Institute IRCCS Foundation, Milan, Italy
| | - Vincent Timmerman
- Peripheral Neuropathy Research Group, University of Antwerp, Antwerp, Belgium.,Institute Born Bunge, Antwerp, Belgium
| |
Collapse
|
6
|
Hensel N, Claus P. The Actin Cytoskeleton in SMA and ALS: How Does It Contribute to Motoneuron Degeneration? Neuroscientist 2017; 24:54-72. [PMID: 28459188 DOI: 10.1177/1073858417705059] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are neurodegenerative diseases with overlapping clinical phenotypes based on impaired motoneuron function. However, the pathomechanisms of both diseases are largely unknown, and it is still unclear whether they converge on the molecular level. SMA is a monogenic disease caused by low levels of functional Survival of Motoneuron (SMN) protein, whereas ALS involves multiple genes as well as environmental factors. Recent evidence argues for involvement of actin regulation as a causative and dysregulated process in both diseases. ALS-causing mutations in the actin-binding protein profilin-1 as well as the ability of the SMN protein to directly bind to profilins argue in favor of a common molecular mechanism involving the actin cytoskeleton. Profilins are major regulat ors of actin-dynamics being involved in multiple neuronal motility and transport processes as well as modulation of synaptic functions that are impaired in models of both motoneuron diseases. In this article, we review the current literature in SMA and ALS research with a focus on the actin cytoskeleton. We propose a common molecular mechanism that explains the degeneration of motoneurons for SMA and some cases of ALS.
Collapse
Affiliation(s)
- Niko Hensel
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany
| | - Peter Claus
- 1 Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany.,2 Niedersachsen Network on Neuroinfectiology (N-RENNT), Hannover, Germany.,3 Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
7
|
Uezu A, Kanak DJ, Bradshaw TWA, Soderblom EJ, Catavero CM, Burette AC, Weinberg RJ, Soderling SH. Identification of an elaborate complex mediating postsynaptic inhibition. Science 2017; 353:1123-9. [PMID: 27609886 DOI: 10.1126/science.aag0821] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
Inhibitory synapses dampen neuronal activity through postsynaptic hyperpolarization. The composition of the inhibitory postsynapse and the mechanistic basis of its regulation, however, remain poorly understood. We used an in vivo chemico-genetic proximity-labeling approach to discover inhibitory postsynaptic proteins. Quantitative mass spectrometry not only recapitulated known inhibitory postsynaptic proteins but also revealed a large network of new proteins, many of which are either implicated in neurodevelopmental disorders or are of unknown function. Clustered regularly interspaced short palindromic repeats (CRISPR) depletion of one of these previously uncharacterized proteins, InSyn1, led to decreased postsynaptic inhibitory sites, reduced the frequency of miniature inhibitory currents, and increased excitability in the hippocampus. Our findings uncover a rich and functionally diverse assemblage of previously unknown proteins that regulate postsynaptic inhibition and might contribute to developmental brain disorders.
Collapse
Affiliation(s)
- Akiyoshi Uezu
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA
| | - Daniel J Kanak
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA
| | - Tyler W A Bradshaw
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA
| | - Erik J Soderblom
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA. Duke Proteomics and Metabolomics Shared Resource and Duke Center for Genomic and Computational Biology, Duke University Medical School, Durham, NC 27703, USA
| | - Christina M Catavero
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA
| | - Alain C Burette
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA. Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Richard J Weinberg
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA. Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott H Soderling
- The Department of Cell Biology, Duke University Medical School, Durham, NC 27703, USA. The Department of Neurobiology, Duke University Medical School, Durham, NC 27703, USA.
| |
Collapse
|
8
|
Hannan MA, Mohibbullah M, Hong YK, Moon IS. Proteomic Analysis of the Neurotrophic Effect of Gelidium amansii in Primary Cultured Neurons. J Med Food 2017; 20:279-287. [DOI: 10.1089/jmf.2016.3848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Md. Abdul Hannan
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Md. Mohibbullah
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Busan, Republic of Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University Graduate School of Medicine, Gyeongju, Republic of Korea
| |
Collapse
|
9
|
Alvarez FJ. Gephyrin and the regulation of synaptic strength and dynamics at glycinergic inhibitory synapses. Brain Res Bull 2016; 129:50-65. [PMID: 27612963 DOI: 10.1016/j.brainresbull.2016.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 08/23/2016] [Accepted: 09/05/2016] [Indexed: 01/23/2023]
Abstract
Glycinergic synapses predominate in brainstem and spinal cord where they modulate motor and sensory processing. Their postsynaptic mechanisms have been considered rather simple because they lack a large variety of glycine receptor isoforms and have relatively simple postsynaptic densities at the ultrastructural level. However, this simplicity is misleading being their postsynaptic regions regulated by a variety of complex mechanisms controlling the efficacy of synaptic inhibition. Early studies suggested that glycinergic inhibitory strength and dynamics depend largely on structural features rather than on molecular complexity. These include regulation of the number of postsynaptic glycine receptors, their localization and the amount of co-localized GABAA receptors and GABA-glycine co-transmission. These properties we now know are under the control of gephyrin. Gephyrin is the first postsynaptic scaffolding protein ever discovered and it was recently found to display a large degree of variation and regulation by splice variants, posttranslational modifications, intracellular trafficking and interactions with the underlying cytoskeleton. Many of these mechanisms are governed by converging excitatory activity and regulate gephyrin oligomerization and receptor binding, the architecture of the postsynaptic density (and by extension the whole synaptic complex), receptor retention and stability. These newly uncovered molecular mechanisms define the size and number of gephyrin postsynaptic regions and the numbers and proportions of glycine and GABAA receptors contained within. All together, they control the emergence of glycinergic synapses of different strength and temporal properties to best match the excitatory drive received by each individual neuron or local dendritic compartment.
Collapse
Affiliation(s)
- Francisco J Alvarez
- Department of Physiology, Emory University, Atlanta, GA 30322-3110, United States.
| |
Collapse
|
10
|
Tariq N, Basharat Z, Butt S, Baig DN. Distribution analysis of profilin isoforms at transcript resolution with mRNA-seq and secondary structure in various organs of Rattus norvegicus. Gene 2016; 589:49-55. [PMID: 27185630 DOI: 10.1016/j.gene.2016.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 11/15/2022]
Abstract
Profilin (Pfn) is an actin binding protein, ubiquitously found in mammals and is essential for the actin polymerization in cells. In brain, it plays a pivotal role in neurogenesis and synapse formation by interacting with various proteins. Four Pfn isoforms have been identified in mammals. This study presents the identification and transcriptional expression of various Pfn isoforms (Pfn1, Pfn2, Pfn3 and Pfn4) in brain, heart, kidney, liver, and muscle and testis of Rattus norvegicus. Organs have been classified into groups based on some similarities. Group I includes brain and testis, Group II includes skeletal muscle and heart, while Group III includes kidney and liver. Pfn1 has been identified in all groups, Pfn2 and Pfn3 have been identified in group I, group III and in one organ (skeletal muscle) of group II. To the best of the authors knowledge, no report of Pfn1 and Pfn2 presence in testis, Pfn3 in brain, liver and skeletal muscle, Pfn4 in kidney and skeletal muscle exists to date. Transcriptional expression showed variations among expression level of different Pfn isoforms in various organs with respect to the control gene GADPH. We hypothesize that this could be attributed to profilin isoform specific mRNA structure and corresponding motifs, which generally contribute to similar or varied decay rates, cellular localization, post transcriptional regulation pattern and ligand binding.
Collapse
Affiliation(s)
- Naila Tariq
- Department of Biological sciences, Forman Christian College (A Chartered University), 54600 Lahore, Pakistan
| | - Zarrin Basharat
- Department of Environmental Sciences, Fatima Jinnah Women University, 46000 Rawalpindi, Pakistan
| | - Saba Butt
- Department of Biological sciences, Forman Christian College (A Chartered University), 54600 Lahore, Pakistan
| | - Deeba Noreen Baig
- Department of Biological sciences, Forman Christian College (A Chartered University), 54600 Lahore, Pakistan
| |
Collapse
|
11
|
Rao R, Ennis K, Mitchell EP, Tran PV, Gewirtz JC. Recurrent Moderate Hypoglycemia Suppresses Brain-Derived Neurotrophic Factor Expression in the Prefrontal Cortex and Impairs Sensorimotor Gating in the Posthypoglycemic Period in Young Rats. Dev Neurosci 2016; 38:74-82. [PMID: 26820887 DOI: 10.1159/000442878] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/29/2015] [Indexed: 01/04/2023] Open
Abstract
Recurrent hypoglycemia is common in infants and children. In developing rat models, recurrent moderate hypoglycemia leads to neuronal injury in the medial prefrontal cortex. To understand the effects beyond neuronal injury, 3-week-old male rats were subjected to 5 episodes of moderate hypoglycemia (blood glucose concentration, approx. 30 mg/dl for 90 min) once daily from postnatal day 24 to 28. Neuronal injury was determined using Fluoro-Jade B histochemistry on postnatal day 29. The effects on brain-derived neurotrophic factor (BDNF) and its cognate receptor, tyrosine kinase receptor B (TrkB) expression, which is critical for prefrontal cortex development, were determined on postnatal day 29 and at adulthood. The effects on prefrontal cortex-mediated function were determined by assessing the prepulse inhibition of the acoustic startle reflex on postnatal day 29 and 2 weeks later, and by testing for fear-potentiated startle at adulthood. Recurrent hypoglycemia led to neuronal injury confined primarily to the medial prefrontal cortex. BDNF/TrkB expression in the prefrontal cortex was suppressed on postnatal day 29 and was accompanied by lower prepulse inhibition, suggesting impaired sensorimotor gating. Following the cessation of recurrent hypoglycemia, the prepulse inhibition had recovered at 2 weeks. BDNF/TrkB expression in the prefrontal cortex had normalized and fear-potentiated startle was intact at adulthood. Recurrent moderate hypoglycemia during development has significant adverse effects on the prefrontal cortex in the posthypoglycemic period.
Collapse
|
12
|
Schweinhuber SK, Meßerschmidt T, Hänsch R, Korte M, Rothkegel M. Profilin isoforms modulate astrocytic morphology and the motility of astrocytic processes. PLoS One 2015; 10:e0117244. [PMID: 25629407 PMCID: PMC4309604 DOI: 10.1371/journal.pone.0117244] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 12/22/2014] [Indexed: 01/17/2023] Open
Abstract
The morphology of astrocytic processes determines their close structural association with synapses referred to as the ‘tripartite synapse’. Concerted morphological plasticity processes at tripartite synapses are supposed to shape neuronal communication. Morphological changes in astrocytes as well as the motility of astrocytic processes require remodeling of the actin cytoskeleton. Among the regulators of fast timescale actin-based motility, the actin binding protein profilin 1 has recently been shown to control the activity-dependent outgrowth of astrocytic processes. Here, we demonstrate that cultured murine astrocytes in addition to the ubiquitous profilin 1 also express the neuronal isoform profilin 2a. To analyze the cellular function of both profilins in astrocytes, we took advantage of a shRNA mediated isoform-specific downregulation. Interestingly, consistent with earlier results in neurons, we found redundant as well as isoform-specific functions of both profilins in modulating cellular physiology. The knockdown of either profilin 1 or profilin 2a led to a significant decrease in cell spreading of astrocytes. In contrast, solely the knockdown of profilin 2a resulted in a significantly reduced morphological complexity of astrocytes in both dissociated and slice culture astrocytes. Moreover, both isoforms proved to be crucial for forskolin-induced astrocytic stellation. Furthermore, forskolin treatment resulted in isoform-specific changes in the phosphorylation level of profilin 1 and profilin 2a, leading to a PKA-dependent phosphorylation of profilin 2a. In addition, transwell assays revealed an involvement of both isoforms in the motility of astrocytic processes, while FRAP analysis displayed an isoform-specific role of profilin 1 in the regulation of actin dynamics in peripheral astrocytic processes. Taken together, we suggest profilin isoforms to be important modulators of astrocytic morphology and motility with overlapping as well as isoform-specific functions.
Collapse
Affiliation(s)
| | - Tania Meßerschmidt
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Robert Hänsch
- Molecular and Cell Biology of Plants, Institute of Plant Biology, TU Braunschweig, Braunschweig, Germany
| | - Martin Korte
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
| | - Martin Rothkegel
- Cellular Neurobiology, Zoological Institute, TU Braunschweig, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
13
|
Abstract
The neurotransmitters GABA and glycine mediate fast synaptic inhibition by activating ligand-gated chloride channels--namely, type A GABA (GABA(A)) and glycine receptors. Both types of receptors are anchored postsynaptically by gephyrin, which self-assembles into a scaffold and interacts with the cytoskeleton. Current research indicates that postsynaptic gephyrin clusters are dynamic assemblies that are held together and regulated by multiple protein-protein interactions. Moreover, post-translational modifications of gephyrin regulate the formation and plasticity of GABAergic synapses by altering the clustering properties of postsynaptic scaffolds and thereby the availability and function of receptors and other signalling molecules. Here, we discuss the formation and regulation of the gephyrin scaffold, its role in GABAergic and glycinergic synaptic function and the implications for the pathophysiology of brain disorders caused by abnormal inhibitory neurotransmission.
Collapse
|
14
|
Choi YN, Lee SK, Seo TW, Lee JS, Yoo SJ. C-terminus of Hsc70-interacting protein regulates profilin1 and breast cancer cell migration. Biochem Biophys Res Commun 2014; 446:1060-6. [DOI: 10.1016/j.bbrc.2014.03.061] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 03/15/2014] [Indexed: 10/25/2022]
|
15
|
Pitychoutis PM, Sanoudou D, Papandreou M, Nasias D, Kouskou M, Tomlinson CR, Tsonis PA, Papadopoulou-Daifoti Z. Forced swim test induces divergent global transcriptomic alterations in the hippocampus of high versus low novelty-seeker rats. Hum Genomics 2014; 8:4. [PMID: 24568636 PMCID: PMC3941591 DOI: 10.1186/1479-7364-8-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/14/2014] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Many neuropsychiatric disorders, including stress-related mood disorders, are complex multi-parametric syndromes. Susceptibility to stress and depression is individually different. The best animal model of individual differences that can be used to study the neurobiology of affect regards spontaneous reactions to novelty. Experimentally, when naive rats are exposed to the stress of a novel environment, they display a highly variable exploratory activity and are classified as high or low responders (HR or LR, respectively). Importantly, HR and LR rats do not seem to exhibit a substantial differentiation in relation to their 'depressive-like' status in the forced swim test (FST), a widely used animal model of 'behavioral despair'. In the present study, we investigated whether FST exposure would be accompanied by phenotype-dependent differences in hippocampal gene expression in HR and LR rats. RESULTS HR and LR rats present a distinct behavioral pattern in the pre-test session but develop comparable depressive-like status in the second FST session. At 24 h following the second FST session, HR and LR rats (stressed and unstressed controls) were sacrificed and hippocampal samples were independently analyzed on whole rat genome Illumina arrays. Functional analysis into pathways and networks was performed using Ingenuity Pathway Analysis (IPA) software. Notably, hippocampal gene expression signatures between HR and LR rats were markedly divergent, despite their comparable depressive-like status in the FST. These molecular differences are reflected in both the extent of transcriptional remodeling (number of significantly changed genes) and the types of molecular pathways affected following FST exposure. A markedly higher number of genes (i.e., 2.28-fold) were statistically significantly changed following FST in LR rats, as compared to their HR counterparts. Notably, genes associated with neurogenesis and synaptic plasticity were induced in the hippocampus of LR rats in response to FST, whereas in HR rats, FST induced pathways directly or indirectly associated with induction of apoptotic mechanisms. CONCLUSIONS The markedly divergent gene expression signatures exposed herein support the notion that the hippocampus of HR and LR rats undergoes distinct transcriptional remodeling in response to the same stress regimen, thus yielding a different FST-related 'endophenotype', despite the seemingly similar depressive-like phenotype.
Collapse
Affiliation(s)
- Pothitos M Pitychoutis
- Department of Biology & Center for Tissue Regeneration and Engineering (TREND), University of Dayton, 300 College Park, Dayton, OH 45469-2320, USA.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Federighi G, Traina G, Macchi M, Ciampini C, Bernardi R, Baldi E, Bucherelli C, Brunelli M, Scuri R. Modulation of gene expression in contextual fear conditioning in the rat. PLoS One 2013; 8:e80037. [PMID: 24278235 PMCID: PMC3837011 DOI: 10.1371/journal.pone.0080037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/27/2013] [Indexed: 01/30/2023] Open
Abstract
In contextual fear conditioning (CFC) a single training leads to long-term memory of context-aversive electrical foot-shocks association. Mid-temporal regions of the brain of trained and naive rats were obtained 2 days after conditioning and screened by two-directional suppression subtractive hybridization. A pool of differentially expressed genes was identified and some of them were randomly selected and confirmed with qRT-PCR assay. These transcripts showed high homology for rat gene sequences coding for proteins involved in different cellular processes. The expression of the selected transcripts was also tested in rats which had freely explored the experimental apparatus (exploration) and in rats to which the same number of aversive shocks had been administered in the same apparatus, but temporally compressed so as to make the association between painful stimuli and the apparatus difficult (shock-only). Some genes resulted differentially expressed only in the rats subjected to CFC, others only in exploration or shock-only rats, whereas the gene coding for translocase of outer mitochondrial membrane 20 protein and nardilysin were differentially expressed in both CFC and exploration rats. For example, the expression of stathmin 1 whose transcripts resulted up regulated was also tested to evaluate the transduction and protein localization after conditioning.
Collapse
Affiliation(s)
- Giuseppe Federighi
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Giovanna Traina
- Dipartimento di Scienze Economico-Estimative e degli Alimenti, Sezione di Chimica Bromatologica, Biochimica, Fisiologia e Nutrizione, Università degli Studi di Perugia, Perugia, Italy
| | - Monica Macchi
- Dipartimento di Biologia, Università di Pisa, Pisa, Italy
| | - Cristina Ciampini
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| | - Rodolfo Bernardi
- Dipartimento di Scienze Agrarie, Genetica Alimentari e Agro-Ambientali, Università di Pisa, Pisa, Italy
| | - Elisabetta Baldi
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | - Corrado Bucherelli
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | | | - Rossana Scuri
- Dipartimento di Ricerca Traslazionale e Delle Nuove Tecnologie in Medicina e Chirurgia, Unità di Fisiologia, Università di Pisa, Pisa, Italy
| |
Collapse
|
17
|
Purkinje cell loss and motor coordination defects in profilin1 mutant mice. Neuroscience 2012; 223:355-64. [PMID: 22864186 DOI: 10.1016/j.neuroscience.2012.07.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 07/05/2012] [Accepted: 07/26/2012] [Indexed: 10/28/2022]
Abstract
Profilin1 is an actin monomer-binding protein, essential for cytoskeletal dynamics. Based on its broad expression in the brain and the localization at excitatory synapses (hippocampal CA3-CA1 synapse, cerebellar parallel fiber (PF)-Purkinje cell (PC) synapse), an important role for profilin1 in brain development and synapse physiology has been postulated. We recently showed normal physiology of hippocampal CA3-CA1 synapses in the absence of profilin1, but impaired glial cell binding and radial migration of cerebellar granule neurons (CGNs). Consequently, brain-specific inactivation of profilin1 by exploiting conditional mutants and Nestin-mediated cre expression resulted in a cerebellar hypoplasia, aberrant organization of cerebellar cortex layers, and ectopic CGNs. Apart from these findings we noted a loss of PCs and an irregularly shaped PC layer in adult mutants. In this study, we show that PC migration and development are not affected in profilin1 mutants, suggesting cell type-specific functions for profilin1 in PCs and CGNs. PC loss begins during the second postnatal week and progresses until adulthood with no further impairment in aged mutants. In Nestin-cre profilin1 mutants, defects in cerebellar cortex cytoarchitecture are associated with impaired motor coordination. However, in L7-cre mutants, lacking profilin1 specifically in PCs, the cerebellar cortex cytoarchitecture is unchanged. Thereby, our results demonstrate that the loss of PCs is not caused by cell-autonomous defects, but presumably by impaired CGN migration. Finally, we show normal functionality of PF-PC synapses in the absence of profilin1. In summary, we conclude that profilin1 is crucially important for brain development, but dispensable for the physiology of excitatory synapses.
Collapse
|