1
|
Markiewicz R, Markiewicz-Gospodarek A, Borowski B, Trubalski M, Łoza B. Reelin Signaling and Synaptic Plasticity in Schizophrenia. Brain Sci 2023; 13:1704. [PMID: 38137152 PMCID: PMC10741648 DOI: 10.3390/brainsci13121704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Recent research emphasizes the significance of studying the quality of life of schizophrenia patients, considering the complex nature of the illness. Identifying neuronal markers for early diagnosis and treatment is crucial. Reelin (RELN) stands out among these markers, with genetic studies highlighting its role in mental health. Suppression of RELN expression may contribute to cognitive deficits by limiting dendritic proliferation, affecting neurogenesis, and leading to improper neuronal circuits. Although the physiological function of reelin is not fully understood, it plays a vital role in hippocampal cell stratification and neuroglia formation. This analysis explores reelin's importance in the nervous system, shedding light on its impact on mental disorders such as schizophrenia, paving the way for innovative therapeutic approaches, and at the same time, raises the following conclusions: increased methylation levels of the RELN gene in patients with a diagnosis of schizophrenia results in a multiple decrease in the expression of reelin, and monitoring of this indicator, i.e., methylation levels, can be used to monitor the severity of symptoms in the course of schizophrenia.
Collapse
Affiliation(s)
- Renata Markiewicz
- Occupational Therapy Laboratory, Chair of Nursing Development, Medical University of Lublin, 4 Staszica St., 20-081 Lublin, Poland;
| | | | - Bartosz Borowski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (B.B.); (M.T.)
| | - Mateusz Trubalski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (B.B.); (M.T.)
| | - Bartosz Łoza
- Department of Psychiatry, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
2
|
Joly-Amado A, Kulkarni N, Nash KR. Reelin Signaling in Neurodevelopmental Disorders and Neurodegenerative Diseases. Brain Sci 2023; 13:1479. [PMID: 37891846 PMCID: PMC10605156 DOI: 10.3390/brainsci13101479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Reelin is an extracellular matrix glycoprotein involved in neuronal migration during embryonic brain development and synaptic plasticity in the adult brain. The role of Reelin in the developing central nervous system has been extensively characterized. Indeed, a loss of Reelin or a disruption in its signaling cascade leads to neurodevelopmental defects and is associated with ataxia, intellectual disability, autism, and several psychiatric disorders. In the adult brain, Reelin is critically involved in neurogenesis and synaptic plasticity. Reelin's signaling potentiates glutamatergic and GABAergic neurotransmission, induces synaptic maturation, and increases AMPA and NMDA receptor subunits' expression and activity. As a result, there is a growing literature reporting that a loss of function and/or reduction of Reelin is implicated in numerous neurodegenerative diseases. The present review summarizes the current state of the literature regarding the implication of Reelin and Reelin-mediated signaling during aging and neurodegenerative disorders, highlighting Reelin as a possible target in the prevention or treatment of progressive neurodegeneration.
Collapse
Affiliation(s)
- Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (N.K.); (K.R.N.)
| | | | | |
Collapse
|
3
|
Lintas C, Cassano I, Azzarà A, Stigliano MG, Gregorj C, Sacco R, Stoccoro A, Coppedè F, Gurrieri F. Maternal Epigenetic Dysregulation as a Possible Risk Factor for Neurodevelopmental Disorders. Genes (Basel) 2023; 14:genes14030585. [PMID: 36980856 PMCID: PMC10048308 DOI: 10.3390/genes14030585] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Neurodevelopmental Disorders (NDs) are a heterogeneous group of disorders and are considered multifactorial diseases with both genetic and environmental components. Epigenetic dysregulation driven by adverse environmental factors has recently been documented in neurodevelopmental disorders as the possible etiological agent for their onset. However, most studies have focused on the epigenomes of the probands rather than on a possible epigenetic dysregulation arising in their mothers and influencing neurodevelopment during pregnancy. The aim of this research was to analyze the methylation profile of four well-known genes involved in neurodevelopment (BDNF, RELN, MTHFR and HTR1A) in the mothers of forty-five age-matched AS (Asperger Syndrome), ADHD (Attention Deficit Hyperactivity Disorder) and typically developing children. We found a significant increase of methylation at the promoter of the RELN and HTR1A genes in AS mothers compared to ADHD and healthy control mothers. For the MTHFR gene, promoter methylation was significantly higher in AS mothers compared to healthy control mothers only. The observed dysregulation in AS mothers could potentially contribute to the affected condition in their children deserving further investigation.
Collapse
Affiliation(s)
- Carla Lintas
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
- Correspondence: ; Tel.: +39-06-225419174
| | - Ilaria Cassano
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Alessia Azzarà
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Maria Grazia Stigliano
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Gregorj
- Operative Research Unit of Hematology, Stem Cell Transplantation, Transfusion Medicine and Cellular Therapy, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Roberto Sacco
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Andrea Stoccoro
- Medical Genetics Laboratory, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Fabio Coppedè
- Medical Genetics Laboratory, Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Fiorella Gurrieri
- Research Unit of Medical Genetics, Department of Medicine, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Operative Research Unit of Medical Genetics, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| |
Collapse
|
4
|
Identification and Validation of RELN Mutation as a Response Indicator for Immune Checkpoint Inhibitor Therapy in Melanoma and Non-Small Cell Lung Cancer. Cells 2022; 11:cells11233841. [PMID: 36497098 PMCID: PMC9741468 DOI: 10.3390/cells11233841] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Remarkable clinical benefits in several advanced cancers are observed under the treatment of immune checkpoint inhibitor (ICI) agents. However, only a smaller proportion of patients respond to the treatments. Reelin (RELN) is frequently mutated in the cancer genome. In this study, the RELN mutation association with ICI treatment efficacy in melanoma and non-small cell lung cancer (NSCLC) was elucidated. Data from 631 melanoma and 109 NSCLC patients with both ICI treatment data and pre-treatment mutational profiles were collected. In addition, from the Cancer Genome Atlas (TCGA) project, we also obtained both tumors to explore the immunologic features behind RELN mutations. Melanoma patients with RELN mutations exhibited a favorable ICI survival benefit when compared with wild-type patients (HR: 0.66, 95% CI: 0.51-0.87, p = 0.003). A higher response rate was also noticed in RELN-mutated patients (38.9% vs. 28.3%, p = 0.017). The association of RELN mutations with a preferable immunotherapy outcome and response was further confirmed in NSCLC. Further exploration demonstrated that favorable immunocyte infiltration and immune response signaling pathways were found in patients with RELN mutations. In this study, RELN mutations were identified to connect with a better immune microenvironment and an improved ICI efficacy in melanoma and NSCLC, which provides a potential biomarker for immunological feature evaluation and immunotherapeutic outcome prediction at the molecular level.
Collapse
|
5
|
Tavernier LJM, Vanpoucke T, Schrauwen I, Van Camp G, Fransen E. Targeted Resequencing of Otosclerosis Patients from Different Populations Replicates Results from a Previous Genome-Wide Association Study. J Clin Med 2022; 11:6978. [PMID: 36498562 PMCID: PMC9737413 DOI: 10.3390/jcm11236978] [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: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Otosclerosis is one of the most common causes of hearing loss in young adults. It has a prevalence of 0.3-0.4% in the European population. Clinical symptoms usually occur between the second and fifth decade of life. Different studies have been performed to unravel the genetic architecture of the disease. Recently, a genome-wide association study (GWAS) identified 15 novel risk loci and replicated the regions of three previously reported candidate genes. In this study, seven candidate genes from the GWAS were resequenced using single molecule molecular inversion probes (smMIPs). smMIPs were used to capture the exonic regions and the 3' and 5' untranslated regions (UTR). Discovered variants were tested for association with the disease using single variant and gene-based association analysis. The single variant results showed that 13 significant variants were associated with otosclerosis. Associated variants were found in five of the seven genes studied here, including AHSG, LINC01482, MARK3, SUPT3H and RELN. Conversely, burden testing did not show a major role of rare variants in the disease. In conclusion, this study was able to replicate five out of seven candidate genes reported in the previous GWAS. This association is likely mainly driven by common variants.
Collapse
Affiliation(s)
- Lisse J. M. Tavernier
- Center of Medical Genetics, University of Antwerp & Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Thomas Vanpoucke
- Center of Medical Genetics, University of Antwerp & Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Isabelle Schrauwen
- Center for Statistical Genetics, Department of Neurology, Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Guy Van Camp
- Center of Medical Genetics, University of Antwerp & Antwerp University Hospital, 2650 Antwerp, Belgium
| | - Erik Fransen
- Center of Medical Genetics, University of Antwerp & Antwerp University Hospital, 2650 Antwerp, Belgium
- StatUA Center for Statistics, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
6
|
Stoyanova II, Lutz D. Functional Diversity of Neuronal Cell Adhesion and Recognition Molecule L1CAM through Proteolytic Cleavage. Cells 2022; 11:cells11193085. [PMID: 36231047 PMCID: PMC9562852 DOI: 10.3390/cells11193085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
The neuronal cell adhesion and recognition molecule L1 does not only 'keep cells together' by way of homophilic and heterophilic interactions, but can also promote cell motility when cleaved into fragments by several proteases. It has largely been thought that such fragments are signs of degradation. Now, it is clear that proteolysis contributes to the pronounced functional diversity of L1, which we have reviewed in this work. L1 fragments generated at the plasma membrane are released into the extracellular space, whereas other membrane-bound fragments are internalised and enter the nucleus, thus conveying extracellular signals to the cell interior. Post-translational modifications on L1 determine the sequence of cleavage by proteases and the subcellular localisation of the generated fragments. Inside the neuronal cells, L1 fragments interact with various binding partners to facilitate morphogenic events, as well as regenerative processes. The stimulation of L1 proteolysis via injection of L1 peptides or proteases active on L1 or L1 mimetics is a promising tool for therapy of injured nervous systems. The collective findings gathered over the years not only shed light on the great functional diversity of L1 and its fragments, but also provide novel mechanistic insights into the adhesion molecule proteolysis that is active in the developing and diseased nervous system.
Collapse
Affiliation(s)
- Irina I. Stoyanova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University, 9002 Varna, Bulgaria
- Department of Brain Ischemia Mechanisms, Research Institute, Medical University, 9002 Varna, Bulgaria
- Correspondence: (I.I.S.); (D.L.)
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum,
44801 Bochum, Germany
- Correspondence: (I.I.S.); (D.L.)
| |
Collapse
|
7
|
Biamonte F, Sica G, Filippini A, D’Alessio A. Evidence of Reelin Signaling in GBM and Its Derived Cancer Stem Cells. Brain Sci 2021; 11:brainsci11060745. [PMID: 34205192 PMCID: PMC8227261 DOI: 10.3390/brainsci11060745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/22/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and malignant form of primary brain cancer, characterized by an overall survival time ranging from 12 to 18 months. Despite the progress in the clinical treatment and the growing number of experimental data aimed at investigating the molecular bases of GBM development, the disease remains characterized by a poor prognosis. Recent studies have proposed the existence of a population of GBM cancer stem cells (CSCs) endowed with self-renewal capability and a high tumorigenic potential that are believed to be responsible for the resistance against common chemotherapy and radiotherapy treatments. Reelin is a large secreted extracellular matrix glycoprotein, which contributes to positioning, migration, and laminar organization of several central nervous system structures during brain development. Mutations of the reelin gene have been linked to disorganization of brain structures during development and behavioral anomalies. In this study, we explored the expression of reelin in GBM and its related peritumoral tissue and performed the same analysis in CSCs isolated from both GBM (GCSCs) and peritumoral tissue (PCSCs) of human patients. Our findings reveal (i) the higher expression of reelin in GBM compared to the peritumoral tissue by immunohistochemical analysis, (ii) the mRNA expression of both reelin and its adaptor molecule Dab1 in either CSC subtypes, although at a different extent; and (iii) the contribution of CSCs-derived reelin in the migration of human primary GBM cell line U87MG. Taken together, our data indicate that the expression of reelin in GBM may represent a potential contribution to the regulation of GBM cancer stem cells behavior, further stimulating the interest on the reelin pathway as a potential target for GBM treatment.
Collapse
Affiliation(s)
- Filippo Biamonte
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
| | - Gigliola Sica
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
| | - Antonio Filippini
- Dipartimento di Scienze Anatomiche, Istologiche, Medico Legali e dell’Apparato Locomotore, Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy;
| | - Alessio D’Alessio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italy;
- Correspondence:
| |
Collapse
|
8
|
Ndoye A, Miskin RP, DiPersio CM. Integrin α3β1 Represses Reelin Expression in Breast Cancer Cells to Promote Invasion. Cancers (Basel) 2021; 13:cancers13020344. [PMID: 33477804 PMCID: PMC7832892 DOI: 10.3390/cancers13020344] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the second leading cause of cancer-related deaths in women, and about 1 in 8 women in the United States develops invasive breast cancer in her lifetime. Integrin α3β1 has been linked to breast cancer progression, but mechanisms whereby it promotes tumor invasion remain unclear. The goal of our study was to determine how α3β1 drives invasion, towards exploiting this integrin as a therapeutic target for breast cancer. We found that α3β1 represses the expression of Reelin, a secreted glycoprotein that inhibits invasion and for which loss of expression is associated with poor prognosis in breast cancer. We also show that increased Reelin expression following RNAi-mediated suppression of α3β1 causes a significant decrease in breast cancer cell invasion. Our findings demonstrate a critical role for α3β1 in promoting cell invasion through repression of Reelin, highlighting the potential value of this integrin as a therapeutic target for breast cancer. Abstract Integrin α3β1, a cell adhesion receptor for certain laminins, is known to promote breast tumor growth and invasion. Our previous gene microarray study showed that the RELN gene, which encodes the extracellular glycoprotein Reelin, was upregulated in α3β1-deficient (i.e., α3 knockdown) MDA-MB-231 cells. In breast cancer, reduced RELN expression is associated with increased invasion and poor prognosis. In this study we demonstrate that α3β1 represses RELN expression to enhance breast cancer cell invasion. RELN mRNA was significantly increased upon RNAi-mediated α3 knockdown in two triple-negative breast cancer cell lines, MDA-MB-231 and SUM159. Modulation of baseline Reelin levels altered invasive potential, where enhanced Reelin expression in MDA-MB-231 cells reduced invasion, while RNAi-mediated suppression of Reelin in SUM159 cells increased invasion. Moreover, treatment of α3β1-expressing MDA-MB-231 cells with culture medium that was conditioned by α3 knockdown MDA-MB-231 cells led to decreased invasion. RNAi-mediated suppression of Reelin in α3 knockdown MDA-MB-231 cells mitigated this effect of conditioned-medium, identifying secreted Reelin as an inhibitor of cell invasion. These results demonstrate a novel role for α3β1 in repressing Reelin in breast cancer cells to promote invasion, supporting this integrin as a potential therapeutic target.
Collapse
Affiliation(s)
- Abibatou Ndoye
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
| | | | - C. Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, 12208 NY, USA;
- Department of Molecular & Cellular Physiology, Albany Medical College, Albany, 12208 NY, USA
- Correspondence:
| |
Collapse
|
9
|
Association Between REELIN Gene Polymorphisms (rs7341475 and rs262355) and Risk of Schizophrenia: an Updated Meta-analysis. J Mol Neurosci 2020; 71:675-690. [PMID: 32889693 DOI: 10.1007/s12031-020-01696-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022]
Abstract
Schizophrenia (SCZ) is a destructive neuropsychiatric illness affecting millions of people worldwide. The correlation between RELN gene polymorphisms and SCZ was investigated by previous researches, though the results remained conflicting. Based on the available studies, we conducted this meta-analysis to provide a more comprehensive outcome on whether the RELN gene polymorphisms (rs7341475 and rs262355) are associated with SCZ. A total of 15 studies with 25,403 subjects (9047 cases and 16,356 controls) retrieved from PubMed, ScienceDirect, EMBASE, Wiley, BMC, Cochrane, Springer, MDPI, SAGE, and Google Scholar up to June 2020 were included. Meta-analysis was performed using Review Manager 5.3. The heterogeneity was checked using I2 statistics and Q-test, whereas publication bias was also measured. The rs7341475 polymorphism showed a significantly lower risk for SCZ for the allele (A vs. G: OR = 0.93, 95%CI = 0.87-0.99), codominant 1 (AG vs. GG: OR = 0.92, 95%CI = 0.85-0.99), dominant model (AA+AG vs. GG: OR = 0.92, 95%CI = 0.86-0.98), and over dominant model (AG vs. AA+GG: OR = 0.92, 95%Cl = 0.86-0.99). The allele, codominant model 1, and dominant models remained statistically significant after the correction of the Bonferroni (p < 0.025). Subgroup analysis confirmed the association of allele and dominant models in the Caucasian after Bonferroni correction. For rs262355 polymorphism, a significantly increased risk of SCZ was found only in Caucasians for codominant 2, dominant, and allele models, but significance exists only for the allele model after Bonferroni correction. Publication bias was found in the case of codominant 2 and recessive models for rs7341475 in the overall population, but this publication was not found after performing the Bonferroni correction or after performing the subgroup analysis. No such publication was found for rs262355. The results suggest that RELN rs7341475 is associated with a lower risk of SCZ in the overall population and Caucasian population, but rs262355 is associated with an increased risk of SCZ only in the Caucasian population.
Collapse
|
10
|
Naseri R, Navabi SJ, Samimi Z, Mishra AP, Nigam M, Chandra H, Olatunde A, Tijjani H, Morais-Urano RP, Farzaei MH. Targeting Glycoproteins as a therapeutic strategy for diabetes mellitus and its complications. Daru 2020; 28:333-358. [PMID: 32006343 PMCID: PMC7095136 DOI: 10.1007/s40199-020-00327-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Glycoproteins are organic compounds formed from proteins and carbohydrates, which are found in many parts of the living systems including the cell membranes. Furthermore, impaired metabolism of glycoprotein components plays the main role in the pathogenesis of diabetes mellitus. The aim of this study is to investigate the influence of glycoprotein levels in the treatment of diabetes mellitus. METHODS All relevant papers in the English language were compiled by searching electronic databases, including Scopus, PubMed and Cochrane library. The keywords of glycoprotein, diabetes mellitus, glycan, glycosylation, and inhibitor were searched until January 2019. RESULTS Glycoproteins are pivotal elements in the regulation of cell proliferation, growth, maturation and signaling pathways. Moreover, they are involved in drug binding, drug transportation, efflux of chemicals and stability of therapeutic proteins. These functions, structure, composition, linkages, biosynthesis, significance and biological effects are discussed as related to their use as a therapeutic strategy for the treatment of diabetes mellitus and its complications. CONCLUSIONS The findings revealed several chemical and natural compounds have significant beneficial effects on glycoprotein metabolism. The comprehension of glycoprotein structure and functions are very essential and inevitable to enhance the knowledge of glycoengineering for glycoprotein-based therapeutics as may be required for the treatment of diabetes mellitus and its associated complications. Graphical abstract.
Collapse
Affiliation(s)
- Rozita Naseri
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Jafar Navabi
- Internal Medicine Department, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, Hemwati Nandan Bahuguna Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand, 246174, India.
| | - Manisha Nigam
- Department of Biochemistry, Hemwati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Harish Chandra
- Department of Microbiology, Gurukul Kangri Vishwavidhyalya, Haridwar, Uttarakhand, 249404, India
| | - Ahmed Olatunde
- Department of Biochemistry, Abubakar Tafawa Balewa University, Bauchi, Nigeria
| | - Habibu Tijjani
- Natural Product Research Laboratory, Department of Biochemistry, Bauchi State University, Gadau, Nigeria
| | - Raquel P Morais-Urano
- Instituto de Química de São Carlos, Universidade de São Paulo, 13560-970, São Carlos, SP, Brasil
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
11
|
Krzyzanowska A, Cabrerizo M, Clascá F, Ramos-Moreno T. Reelin Immunoreactivity in the Adult Spinal Cord: A Comparative Study in Rodents, Carnivores, and Non-human Primates. Front Neuroanat 2020; 13:102. [PMID: 31969808 PMCID: PMC6960112 DOI: 10.3389/fnana.2019.00102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/12/2019] [Indexed: 11/17/2022] Open
Abstract
Reelin is a large extracellular matrix (ECM) glycoprotein secreted by several neuronal populations in a specific manner in both the developing and the adult central nervous system. The extent of Reelin protein distribution and its functional role in the adult neocortex is well documented in different mammal models. However, its role in the adult spinal cord has not been well characterized and its distribution in the rodent spinal cord is fragmentary and has not been investigated in carnivores or primates as of yet. To gain insight into which neuronal populations and specific circuits may be influenced by Reelin in the adult spinal cord, we have conducted light and confocal microscopy study analysis of Reelin-immunoreactive cell types in the adult spinal cord. Here, we describe and compare Reelin immunoreactive cell type and distribution in the spinal cord of adult non-human primate (macaque monkeys, Macaca mulatta), carnivore (ferret, Mustela putorius) and rodent (rat, Rattus norvegicus). Our results show that in all three species studied, Reelin-immunoreactive neurons are present in the intermediate gray matter, ventricular zone and superficial dorsal horn and intermedio-lateral nucleus, while positive cells in the Clarke nucleus are only found in rats and primates. In addition, Reelin intermediolateral neurons colocalize with choline acetyltransferase (ChAT) only in macaque whilst motor neurons also colocalize Reelin and ChAT in macaque, ferret and rat spinal cord. The different expression patterns might reflect a differential role for Reelin in the pathways involved in the coordination of locomotor activity in the fore- and hind limbs.
Collapse
Affiliation(s)
- Agnieszka Krzyzanowska
- Department of Anatomy and Neuroscience, School of Medicine, Autonoma University, Madrid, Spain.,Division of Urological Cancers, Faculty of Medicine, Lund University, Lund, Sweden
| | - Marina Cabrerizo
- Department of Anatomy and Neuroscience, School of Medicine, Autonoma University, Madrid, Spain.,Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Clascá
- Department of Anatomy and Neuroscience, School of Medicine, Autonoma University, Madrid, Spain
| | - Tania Ramos-Moreno
- Department of Anatomy and Neuroscience, School of Medicine, Autonoma University, Madrid, Spain.,Lund Stem Cell Center, Division of Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Mikulska-Ruminska K, Strzelecki J, Nowak W. Dynamics, nanomechanics and signal transduction in reelin repeats. Sci Rep 2019; 9:18974. [PMID: 31831824 PMCID: PMC6908669 DOI: 10.1038/s41598-019-55461-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/27/2019] [Indexed: 12/04/2022] Open
Abstract
Reelin is a large glycoprotein controlling brain development and cell adhesion. It regulates the positioning of neurons, as well as neurotransmission and memory formation. Perturbations in reelin signaling are linked to psychiatric disorders. Reelin participates in signal transduction by binding to the lipoprotein receptors VLDLR and ApoER2 through its central region. This part is rich in repeating BNR-EGF-BNR modules. We used standard molecular dynamics, steered molecular dynamics, and perturbation response scanning computational methods to characterize unique dynamical properties of reelin modules involved in signaling. Each module has specific sensors and effectors arranged in a similar topology. In the modules studied, disulfide bridges play a protective role, probably making both selective binding and protease activity of reelin possible. Results of single reelin molecule stretching by atomic force microscopy provide the first data on the mechanical stability of individual reelin domains. The forces required for partial unfolding of the modules studied are below 60 pN.
Collapse
Affiliation(s)
- Karolina Mikulska-Ruminska
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland.
| | - Janusz Strzelecki
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland
| | - Wieslaw Nowak
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University, Grudziadzka 5, 87-100, Torun, Poland.
| |
Collapse
|
13
|
Abstract
The complexity of morphogenesis poses a fundamental challenge to understanding the mechanisms governing the formation of biological patterns and structures. Over the past century, numerous processes have been identified as critically contributing to morphogenetic events, but the interplay between the various components and aspects of pattern formation have been much harder to grasp. The combination of traditional biology with mathematical and computational methods has had a profound effect on our current understanding of morphogenesis and led to significant insights and advancements in the field. In particular, the theoretical concepts of reaction–diffusion systems and positional information, proposed by Alan Turing and Lewis Wolpert, respectively, dramatically influenced our general view of morphogenesis, although typically in isolation from one another. In recent years, agent-based modeling has been emerging as a consolidation and implementation of the two theories within a single framework. Agent-based models (ABMs) are unique in their ability to integrate combinations of heterogeneous processes and investigate their respective dynamics, especially in the context of spatial phenomena. In this review, we highlight the benefits and technical challenges associated with ABMs as tools for examining morphogenetic events. These models display unparalleled flexibility for studying various morphogenetic phenomena at multiple levels and have the important advantage of informing future experimental work, including the targeted engineering of tissues and organs.
Collapse
|
14
|
Ducharme P, Zarruk JG, David S, Paquin J. The ferroxidase ceruloplasmin influences Reelin processing, cofilin phosphorylation and neuronal organization in the developing brain. Mol Cell Neurosci 2018; 92:104-113. [PMID: 30077770 DOI: 10.1016/j.mcn.2018.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/10/2018] [Accepted: 07/30/2018] [Indexed: 12/09/2022] Open
Abstract
Ceruloplasmin (Cp) is an important extracellular regulator of iron metabolism. We showed previously that it stimulates Reelin proteolytic processing and cell aggregation in cultures of developing neurons. Reelin is a secreted protein required for the correct positioning of neurons in the brain. It is cleaved in vivo into N-terminally-derived 300K and 180K fragments through incompletely known mechanisms. One of Reelin signaling targets is the actin-binding protein cofilin, the phosphorylation of which is diminished in Reelin-deficient mice. This work looked for in vivo evidence of a relationship between Cp, Reelin and neuronal organization during brain development by analyzing wild-type and Cp-null mice. Cp as well as the full-length, 300K and 180K Reelin species appeared together in wild-type brains at embryonic day (E) 12.5 by immunoblotting. In wild-type compared to Cp-null brains, there was more 300K Reelin from E12.5 to E17.5, a period characterized by extensive, radially directed neuronal migration in the cerebral cortex. Immunofluorescence labeling of tissue sections at E16.5 revealed the localization of Cp with radial glia and meningeal cells adjacent to Reelin-producing Cajal-Retzius neurons, underlining the proximity of Cp and Reelin. Cofilin phosphorylation was seen starting at E10.5-E12.5 and lasted longer until postnatal day 7 in wild-type than Cp-null mice. Finally, using CUX1 as a marker revealed defective accumulation of neurons in layers II/III in neonatal and adult Cp-null mice. These results combined with our earlier work point to a potentially new role of Cp in Reelin processing and signaling and neuronal organization in the cerebral cortex in vivo.
Collapse
Affiliation(s)
- Philippe Ducharme
- Département de Chimie and Centre BioMed, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, Quebec H3C 3P8, Canada.
| | - Juan G Zarruk
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, 1650 Cedar Ave., Montreal, Quebec H3G 1A4, Canada.
| | - Samuel David
- Centre for Research in Neuroscience, The Research Institute of the McGill University Health Center, 1650 Cedar Ave., Montreal, Quebec H3G 1A4, Canada.
| | - Joanne Paquin
- Département de Chimie and Centre BioMed, Université du Québec à Montréal, C.P. 8888, Succ. Centre-ville, Montreal, Quebec H3C 3P8, Canada.
| |
Collapse
|
15
|
Ohja K, Gozal E, Fahnestock M, Cai L, Cai J, Freedman JH, Switala A, El-Baz A, Barnes GN. Neuroimmunologic and Neurotrophic Interactions in Autism Spectrum Disorders: Relationship to Neuroinflammation. Neuromolecular Med 2018; 20:161-173. [PMID: 29691724 PMCID: PMC5942347 DOI: 10.1007/s12017-018-8488-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022]
Abstract
Autism spectrum disorders (ASD) are the most prevalent set of pediatric neurobiological disorders. The etiology of ASD has both genetic and environmental components including possible dysfunction of the immune system. The relationship of the immune system to aberrant neural circuitry output in the form of altered behaviors and communication characterized by ASD is unknown. Dysregulation of neurotrophins such as BDNF and their signaling pathways have been implicated in ASD. While abnormal cortical formation and autistic behaviors in mouse models of immune activation have been described, no one theory has been described to link activation of the immune system to specific brain signaling pathways aberrant in ASD. In this paper we explore the relationship between neurotrophin signaling, the immune system and ASD. To this effect we hypothesize that an interplay of dysregulated immune system, synaptogenic growth factors and their signaling pathways contribute to the development of ASD phenotypes.
Collapse
Affiliation(s)
- Kshama Ohja
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Evelyne Gozal
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Lu Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jun Cai
- Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jonathan H Freedman
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Andy Switala
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, Louisville, KY, USA
| | - Gregory Neal Barnes
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA. .,Department of Pediatrics, University of Louisville School of Medicine, Louisville, KY, USA. .,Spafford Ackerly Chair in Child and Adolescent Psychiatry, University of Louisville Autism Center, 1405 East Burnett Avenue, Louisville, KY, 40217, USA.
| |
Collapse
|
16
|
Shen L, Zhang K, Feng C, Chen Y, Li S, Iqbal J, Liao L, Zhao Y, Zhai J. iTRAQ-Based Proteomic Analysis Reveals Protein Profile in Plasma from Children with Autism. Proteomics Clin Appl 2018; 12:e1700085. [PMID: 29274201 DOI: 10.1002/prca.201700085] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 11/26/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Autism is a childhood neurological disorder with poorly understood etiology and pathology. This study is designed to identify differentially expressed proteins that might serve as potential biomarkers for autism. EXPERIMENTAL DESIGN We perform iTRAQ (isobaric tags for relative and absolute quantitation) analysis for normal and autistic children's plasma of the same age group. RESULTS The results show that 24 differentially expressed proteins were identified between autistic subjects and controls. For the first time, differential expression of complement C5 (C5) and fermitin family homolog 3 (FERMT3) are related to autism. Five proteins, that is, complement C3 (C3), C5, integrin alpha-IIb (ITGA2B), talin-1 (TLN1), and vitamin D-binding protein (GC) were validated via enzyme-linked immunosorbent assay (ELISA). By ROC (receiver operating characteristic) analysis, combinations of these five proteins C3, C5, GC, ITGA2B, and TLN1 distinguished autistic children from healthy controls with a high AUC (area under the ROC curve) value (0.982, 95% CI, 0.957-1.000, p < 0.000). CONCLUSION These above described proteins are found involved in different pathways that have previously been linked to the pathophysiology of autism spectrum disorders (ASDs). The results strongly support that focal adhesions, acting cytoskeleton, cell adhesion, motility and migration, synaptogenesis, and complement system are involved in the pathogenesis of autism, and highlight the important role of platelet function in the pathophysiology of autism.
Collapse
Affiliation(s)
- Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Kaoyuan Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Chengyun Feng
- Maternal and Child Health Hospital of Baoan, Shenzhen, P. R. China
| | - Youjiao Chen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China.,Xiang Ya Changde Hospital, Changde City, Hunan Province, P. R. China
| | - Shuiming Li
- College of Life Science and Oceanography, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen, P. R. China
| | - Javed Iqbal
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Liping Liao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen, P. R. China
| | - Jian Zhai
- Maternal and Child Health Hospital of Baoan, Shenzhen, P. R. China
| |
Collapse
|
17
|
Lutz D, Sharaf A, Drexler D, Kataria H, Wolters-Eisfeld G, Brunne B, Kleene R, Loers G, Frotscher M, Schachner M. Proteolytic cleavage of transmembrane cell adhesion molecule L1 by extracellular matrix molecule Reelin is important for mouse brain development. Sci Rep 2017; 7:15268. [PMID: 29127326 PMCID: PMC5681625 DOI: 10.1038/s41598-017-15311-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/25/2017] [Indexed: 02/05/2023] Open
Abstract
The cell adhesion molecule L1 and the extracellular matrix protein Reelin play crucial roles in the developing nervous system. Reelin is known to activate signalling cascades regulating neuronal migration by binding to lipoprotein receptors. However, the interaction of Reelin with adhesion molecules, such as L1, has remained poorly explored. Here, we report that full-length Reelin and its N-terminal fragments N-R2 and N-R6 bind to L1 and that full-length Reelin and its N-terminal fragment N-R6 proteolytically cleave L1 to generate an L1 fragment with a molecular mass of 80 kDa (L1-80). Expression of N-R6 and generation of L1-80 coincide in time at early developmental stages of the cerebral cortex. Reelin-mediated generation of L1-80 is involved in neurite outgrowth and in stimulation of migration of cultured cortical and cerebellar neurons. Morphological abnormalities in layer formation of the cerebral cortex of L1-deficient mice partially overlap with those of Reelin-deficient reeler mice. In utero electroporation of L1-80 into reeler embryos normalised the migration of cortical neurons in reeler embryos. The combined results indicate that the direct interaction between L1 and Reelin as well as the Reelin-mediated generation of L1-80 contribute to brain development at early developmental stages.
Collapse
Affiliation(s)
- David Lutz
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany. .,Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Ahmed Sharaf
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Dagmar Drexler
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Bianka Brunne
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gabriele Loers
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA. .,Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guandong, 515041, China.
| |
Collapse
|
18
|
Lee JY, Kim MJ, Li L, Velumian AA, Aui PM, Fehlings MG, Petratos S. Nogo receptor 1 regulates Caspr distribution at axo-glial units in the central nervous system. Sci Rep 2017; 7:8958. [PMID: 28827698 PMCID: PMC5567129 DOI: 10.1038/s41598-017-09405-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/24/2017] [Indexed: 11/09/2022] Open
Abstract
Axo-glial units are highly organised microstructures propagating saltatory conduction and are disrupted during multiple sclerosis (MS). Nogo receptor 1 (NgR1) has been suggested to govern axonal damage during the progression of disease in the MS-like mouse model, experimental autoimmune encephalomyelitis (EAE). Here we have identified that adult ngr1 -/- mice, previously used in EAE and spinal cord injury experiments, display elongated paranodes, and nodes of Ranvier. Unstructured paranodal regions in ngr1 -/- mice are matched with more distributed expression pattern of Caspr. Compound action potentials of optic nerves and spinal cords from naïve ngr1 -/- mice are delayed and reduced. Molecular interaction studies revealed enhanced Caspr cleavage. Our data suggest that NgR1 may regulate axo-myelin ultrastructure through Caspr-mediated adhesion, regulating the electrophysiological signature of myelinated axons of central nervous system (CNS).
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Min Joung Kim
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Lijun Li
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexander A Velumian
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pei Mun Aui
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Michael G Fehlings
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia.
| |
Collapse
|
19
|
Carotti S, Perrone G, Amato M, Vespasiani Gentilucci U, Righi D, Francesconi M, Pellegrini C, Zalfa F, Zingariello M, Picardi A, Onetti Muda A, Morini S. Reelin expression in human liver of patients with chronic hepatitis C infection. Eur J Histochem 2017; 61:2745. [PMID: 28348420 PMCID: PMC5365015 DOI: 10.4081/ejh.2017.2745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 02/08/2023] Open
Abstract
Reelin is a secreted extracellular glycoprotein that plays a critical role during brain development. Several studies have described Reelin expression in hepatic stellate cells of the human liver. In order to investigate the possible role of Reelin in the process of hepatic fibrogenesis, in this study we investigated Reelin expression in the liver tissue of patients infected with the Hepatitis C Virus (HCV). On this basis, Reelin expression was analysed by immunohistochemistry during liver biopsies of 81 patients with HCV-related chronic hepatitis. A Knodell score was used to stage liver fibrosis. Hepatic stellate cells/myofibroblast immunohistochemical markers (CRBP-1, alpha-SMA) were also evaluated. As further confirmed by co-localization experiments (Reelin +CRBP-1), Reelin protein was expressed by hepatic stellate cells/myofibroblasts, and a significant positive correlation was found between Reelin expression and the stage of liver fibrosis (P=0.002). Moreover, Reelin correlated with CRBP-1 positive cells (P=0.002), but not with alpha-SMA, suggesting that Reelin should not be regarded as a marker of hepatic stellate cells/myofibroblasts differentiation but rather as a functional protein expressed during some phases of liver fibrosis. Furthermore, Disabled-1 (Dab1), a Reelin adaptor protein, was expressed in cells of ductular reaction suggesting a paracrine role for Reelin with regards these elements. In conclusion, Reelin was expressed by human hepatic stellate cells/myofibroblasts and the number of these cells increased significantly in the lobule as the liver fibrosis progressed, suggesting a role for Reelin in the activation of hepatic stellate cells/myofibroblasts during liver injury. Reelin may potentially be incorporated into liver injury evaluations in combination with other histological data.
Collapse
Affiliation(s)
- Simone Carotti
- Campus Bio-Medico University, Laboratory of Microscopic and Ultrastructural Anatomy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kelemenova S, Ostatnikova D. Androgens Contribute to the Process of Neuronal Development: Implications in Explanation of Autism Pathogenesis. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/bf03379917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abstract
Fetal testosterone significantly influences the brain development. It affects number of neurons and conformation of dendritic spines within the sexual dimorphic preoptic area in the hypothalamus. Excessive testosterone levels in utero possibly contribute to the masculinization of the brain. Evidences of these facts are plausible in the anatomic field as well as behavioral effects both in rat models and in humans. Rats exposed to excessive testosterone doses in utero show masculinized brain anatomy and behavior, such as better spatial visualization performance typical for males. In humans, congenital adrenal hyperplasia that causes elevated androgen level possibly results in masculinized behavior observed in these individuals. There are reasons for the theory of the connection existence between testosterone influence on the brain functions and the pathogenesis of neurodevelopmental disorders. In this review, pathogenesis of autism, the most genetic neurodevelopmental disease is discussed. Autism is a disease with broad genetic heterogeneity and polygenic inheritance. Autism associated genes are localized throughout the genome, with the chromosome 7q most frequently involved. One of these genes encodes reelin protein that is crucial for neuronal migration in the developing brain. The connection between androgens, neuronal migration and neurodevelopmental disorder pathophysiology is also discussed.
Collapse
|
21
|
Lintas C, Sacco R, Persico AM. Differential methylation at the RELN gene promoter in temporal cortex from autistic and typically developing post-puberal subjects. J Neurodev Disord 2016; 8:18. [PMID: 27134686 PMCID: PMC4850686 DOI: 10.1186/s11689-016-9151-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/12/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Reelin plays a pivotal role in neurodevelopment and in post-natal synaptic plasticity and has been implicated in the pathogenesis of autism spectrum disorder (ASD). The reelin (RELN) gene expression is significantly decreased in ASD, both in the brain and peripherally. Methylation at the RELN gene promoter is largely triggered at puberty, and hypermethylation has been found in post-mortem brains of schizophrenic and bipolar patients. METHODS In this study, we assessed RELN gene methylation status in post-mortem temporocortical tissue samples (BA41/42 or 22) of six pairs of post-puberal individuals with ASD and typically developing subjects, matched for sex (male:female, M:F = 5:1), age, and post-mortem interval. RESULTS ASD patients display a significantly higher number of methylated CpG islands and heavier methylation in the 5' region of the RELN gene promoter, spanning from -458 to -223 bp, whereas controls have more methylated CpG positions and greater extent of methylation at the 3' promoter region, spanning from -222 to +1 bp. The most upstream promoter region (-458 to -364 bp) is methylated only in ASD brains, while the most downstream region (-131 to +1 bp) is methylated exclusively in control brains. Within this general framework, three different methylation patterns are discernible, each correlated with different extents of reduction in reelin gene expression among ASD individuals compared to controls. CONCLUSIONS The methylation pattern is different in ASD and control post-mortem brains. ASD-specific CpG positions, located in the most upstream gene promoter region, may exert a functional role potentially conferring ASD risk by blunting RELN gene expression.
Collapse
Affiliation(s)
- Carla Lintas
- Unit of Child and Adolescent Neuropsychiatry, University Campus Bio-Medico, Rome, Italy ; Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Roberto Sacco
- Unit of Child and Adolescent Neuropsychiatry, University Campus Bio-Medico, Rome, Italy ; Laboratory of Molecular Psychiatry and Neurogenetics, University Campus Bio-Medico, Rome, Italy
| | - Antonio M Persico
- Unit of Child and Adolescent Neuropsychiatry, "Gaetano Martino" University Hospital, University of Messina, via Consolare Valeria 1, I-98125 Messina, Italy ; Mafalda Luce Center for Pervasive Developmental Disorders, Milan, Italy
| |
Collapse
|
22
|
Chmielewski WX, Beste C. Action control processes in autism spectrum disorder – Insights from a neurobiological and neuroanatomical perspective. Prog Neurobiol 2015; 124:49-83. [DOI: 10.1016/j.pneurobio.2014.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 12/22/2022]
|
23
|
Hawthorne AL. Repurposing Reelin: the new role of radial glia, Reelin and Notch in motor neuron migration. Exp Neurol 2014; 256:17-20. [PMID: 24607503 DOI: 10.1016/j.expneurol.2014.02.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 01/10/2023]
Abstract
The role of Reelin during cerebral cortical neuron migration has long been studied, but the Reelin signaling pathway and its possible interactions are just beginning to be unraveled. Reelin is not only important in cerebral cortical migration, but has recently been shown to interact with the Notch signaling pathway and to be critical for radial glial cell number and morphology. Lee and Song (2013) show a new Notch- and Reelin-dependent role for radial glia in the mouse spinal cord: to act as a fine filter that allows somatic motor neuron axons but not cell bodies to traverse out of the CNS. Here, the types of neuronal migration are discussed, focusing on motor neurons and cues for proper localization. The interaction of Reelin signaling with the Notch pathway is reviewed, which dictates the proper formation of radial glia in the spinal cord in order to prevent ectopic motor neuron migration (Lee and Song, 2013). Future studies may reveal novel interactions and further insights as to how Reelin functions throughout the developing nervous system.
Collapse
Affiliation(s)
- Alicia L Hawthorne
- Dept. of Cell Biology, Emory University School of Medicine, 615 Michael St., WBRB 415, Atlanta, GA 30322, USA.
| |
Collapse
|
24
|
Rossignol DA, Genuis SJ, Frye RE. Environmental toxicants and autism spectrum disorders: a systematic review. Transl Psychiatry 2014; 4:e360. [PMID: 24518398 PMCID: PMC3944636 DOI: 10.1038/tp.2014.4] [Citation(s) in RCA: 302] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 12/15/2013] [Accepted: 01/06/2014] [Indexed: 11/21/2022] Open
Abstract
Although the involvement of genetic abnormalities in autism spectrum disorders (ASD) is well-accepted, recent studies point to an equal contribution by environmental factors, particularly environmental toxicants. However, these toxicant-related studies in ASD have not been systematically reviewed to date. Therefore, we compiled publications investigating potential associations between environmental toxicants and ASD and arranged these publications into the following three categories: (a) studies examining estimated toxicant exposures in the environment during the preconceptional, gestational and early childhood periods; (b) studies investigating biomarkers of toxicants; and (c) studies examining potential genetic susceptibilities to toxicants. A literature search of nine electronic scientific databases through November 2013 was performed. In the first category examining ASD risk and estimated toxicant exposures in the environment, the majority of studies (34/37; 92%) reported an association. Most of these studies were retrospective case-control, ecological or prospective cohort studies, although a few had weaker study designs (for example, case reports or series). Toxicants implicated in ASD included pesticides, phthalates, polychlorinated biphenyls (PCBs), solvents, toxic waste sites, air pollutants and heavy metals, with the strongest evidence found for air pollutants and pesticides. Gestational exposure to methylmercury (through fish exposure, one study) and childhood exposure to pollutants in water supplies (two studies) were not found to be associated with ASD risk. In the second category of studies investigating biomarkers of toxicants and ASD, a large number was dedicated to examining heavy metals. Such studies demonstrated mixed findings, with only 19 of 40 (47%) case-control studies reporting higher concentrations of heavy metals in blood, urine, hair, brain or teeth of children with ASD compared with controls. Other biomarker studies reported that solvent, phthalate and pesticide levels were associated with ASD, whereas PCB studies were mixed. Seven studies reported a relationship between autism severity and heavy metal biomarkers, suggesting evidence of a dose-effect relationship. Overall, the evidence linking biomarkers of toxicants with ASD (the second category) was weaker compared with the evidence associating estimated exposures to toxicants in the environment and ASD risk (the first category) because many of the biomarker studies contained small sample sizes and the relationships between biomarkers and ASD were inconsistent across studies. Regarding the third category of studies investigating potential genetic susceptibilities to toxicants, 10 unique studies examined polymorphisms in genes associated with increased susceptibilities to toxicants, with 8 studies reporting that such polymorphisms were more common in ASD individuals (or their mothers, 1 study) compared with controls (one study examined multiple polymorphisms). Genes implicated in these studies included paraoxonase (PON1, three of five studies), glutathione S-transferase (GSTM1 and GSTP1, three of four studies), δ-aminolevulinic acid dehydratase (one study), SLC11A3 (one study) and the metal regulatory transcription factor 1 (one of two studies). Notably, many of the reviewed studies had significant limitations, including lack of replication, limited sample sizes, retrospective design, recall and publication biases, inadequate matching of cases and controls, and the use of nonstandard tools to diagnose ASD. The findings of this review suggest that the etiology of ASD may involve, at least in a subset of children, complex interactions between genetic factors and certain environmental toxicants that may act synergistically or in parallel during critical periods of neurodevelopment, in a manner that increases the likelihood of developing ASD. Because of the limitations of many of the reviewed studies, additional high-quality epidemiological studies concerning environmental toxicants and ASD are warranted to confirm and clarify many of these findings.
Collapse
Affiliation(s)
- D A Rossignol
- Family Medicine, Rossignol Medical Center, Irvine, CA, USA
| | - S J Genuis
- Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - R E Frye
- Arkansas Children's Hospital Research Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
25
|
Persico AM, Merelli S. Environmental Factors in the Onset of Autism Spectrum Disorder. CURRENT DEVELOPMENTAL DISORDERS REPORTS 2014. [DOI: 10.1007/s40474-013-0002-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
26
|
KIM JIEUN, SHIN MALSOON, SEO TAEBEOM, JI EUNSANG, BAEK SEONGSOO, LEE SAMJUN, PARK JOONKI, KIM CHANGJU. Treadmill exercise ameliorates motor disturbance through inhibition of apoptosis in the cerebellum of valproic acid-induced autistic rat pups. Mol Med Rep 2013; 8:327-34. [DOI: 10.3892/mmr.2013.1518] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Accepted: 05/20/2013] [Indexed: 11/06/2022] Open
|
27
|
Kneeland RE, Fatemi SH. Viral infection, inflammation and schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2013; 42:35-48. [PMID: 22349576 PMCID: PMC3408569 DOI: 10.1016/j.pnpbp.2012.02.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/06/2012] [Accepted: 02/02/2012] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a severe neurodevelopmental disorder with genetic and environmental etiologies. Prenatal viral/bacterial infections and inflammation play major roles in the genesis of schizophrenia. In this review, we describe a viral model of schizophrenia tested in mice whereby the offspring of mice prenatally infected with influenza at E7, E9, E16, and E18 show significant gene, protein, and brain structural abnormalities postnatally. Similarly, we describe data on rodents exposed to bacterial infection or injected with a synthetic viral mimic (PolyI:C) also demonstrating brain structural and behavioral abnormalities. Moreover, human serologic data has been indispensible in supporting the viral theory of schizophrenia. Individuals born seropositive for bacterial and viral agents are at a significantly elevated risk of developing schizophrenia. While the specific mechanisms of prenatal viral/bacterial infections and brain disorder are unclear, recent findings suggest that the maternal inflammatory response may be associated with fetal brain injury. Preventive and therapeutic treatment options are also proposed. This review presents data related to epidemiology, human serology, and experimental animal models which support the viral model of schizophrenia.
Collapse
Affiliation(s)
- Rachel E. Kneeland
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St. SE, MMC 392, Minneapolis, MN 55455, United States
| | - S. Hossein Fatemi
- Department of Psychiatry, Division of Neuroscience Research, University of Minnesota Medical School, 420 Delaware St. SE, MMC 392, Minneapolis, MN 55455, United States,Department of Pharmacology, University of Minnesota Medical School, 310 Church St. SE, Minneapolis, MN 55455, United States and Department of Neuroscience, University of Minnesota Medical School, 310 Church St. SE, Minneapolis, MN 55455, United States,Corresponding author at: 420 Delaware Street SE, MMC 392, Minneapolis, MN 55455. Tel.: +1 612 626 3633; fax: +1 612 624 8935. (R.E. Kneeland), (S.H. Fatemi)
| |
Collapse
|
28
|
Li M, Luo XJ, Xiao X, Shi L, Liu XY, Yin LD, Ma XY, Yang SY, Pu XF, Yu J, Diao HB, Shi H, Su B. Analysis of common genetic variants identifies RELN as a risk gene for schizophrenia in Chinese population. World J Biol Psychiatry 2013; 14:91-9. [PMID: 21745129 DOI: 10.3109/15622975.2011.587891] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
UNLABELLED Abstract Objectives. Several lines of evidence have shown that both RELN mRNA and protein are possibly down-regulated in the brain of schizophrenia patients. Recent association studies in European populations suggested RELN as a risk gene for schizophrenia. In this study, we test if RELN contributes to the risk of schizophrenia in Chinese population. Methods. We conducted case-control association analysis of 19 representative single nucleotide polymorphisms (SNPs) spanning the entire region of RELN in two independent Han Chinese samples from southwestern China (the Kunming sample and the Yuxi sample). Results. We identified six SNPs significantly associated with schizophrenia in the Kunming sample and four of them remained significant in the combined samples (the P values range from 0.006 to 4.0 × 10(-5)). Haplotype analysis also suggested significant associations for the haplotypes incorporating the six significant SNPs (global P < 1.0 × 10(-5)). Additionally, we also observed several other haplotypes (defined by a different set of SNPs) significantly associated with schizophrenia in the Kunming sample. However, the reported association of rs7341475 in Ashkenazi Jews was not significant in Han Chinese. CONCLUSIONS Our findings demonstrate that RELN is a susceptibility gene for schizophrenia in Chinese population, and it is likely a common risk gene for schizophrenia in major populations worldwide.
Collapse
Affiliation(s)
- Ming Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Stamou M, Streifel KM, Goines PE, Lein PJ. Neuronal connectivity as a convergent target of gene × environment interactions that confer risk for Autism Spectrum Disorders. Neurotoxicol Teratol 2013; 36:3-16. [PMID: 23269408 PMCID: PMC3610799 DOI: 10.1016/j.ntt.2012.12.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 11/12/2012] [Accepted: 12/17/2012] [Indexed: 11/21/2022]
Abstract
Evidence implicates environmental factors in the pathogenesis of Autism Spectrum Disorders (ASD). However, the identity of specific environmental chemicals that influence ASD risk, severity or treatment outcome remains elusive. The impact of any given environmental exposure likely varies across a population according to individual genetic substrates, and this increases the difficulty of identifying clear associations between exposure and ASD diagnoses. Heritable genetic vulnerabilities may amplify adverse effects triggered by environmental exposures if genetic and environmental factors converge to dysregulate the same signaling systems at critical times of development. Thus, one strategy for identifying environmental risk factors for ASD is to screen for environmental factors that modulate the same signaling pathways as ASD susceptibility genes. Recent advances in defining the molecular and cellular pathology of ASD point to altered patterns of neuronal connectivity in the developing brain as the neurobiological basis of these disorders. Studies of syndromic ASD and rare highly penetrant mutations or CNVs in ASD suggest that ASD risk genes converge on several major signaling pathways linked to altered neuronal connectivity in the developing brain. This review briefly summarizes the evidence implicating dysfunctional signaling via Ca(2+)-dependent mechanisms, extracellular signal-regulated kinases (ERK)/phosphatidylinositol-3-kinases (PI3K) and neuroligin-neurexin-SHANK as convergent molecular mechanisms in ASD, and then discusses examples of environmental chemicals for which there is emerging evidence of their potential to interfere with normal neuronal connectivity via perturbation of these signaling pathways.
Collapse
Affiliation(s)
- Marianna Stamou
- Department of Molecular Biosciences, University of California at Davis School of Veterinary Medicine, Davis CA, 95616, United States
| | - Karin M. Streifel
- Department of Molecular Biosciences, University of California at Davis School of Veterinary Medicine, Davis CA, 95616, United States
| | - Paula E. Goines
- Department of Molecular Biosciences, University of California at Davis School of Veterinary Medicine, Davis CA, 95616, United States
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California at Davis School of Veterinary Medicine, Davis CA, 95616, United States
| |
Collapse
|
30
|
Iwata K, Izumo N, Matsuzaki H, Manabe T, Ishibashi Y, Ichitani Y, Yamada K, Thanseem I, Anitha A, Vasu MM, Shimmura C, Wakuda T, Kameno Y, Takahashi T, Iwata Y, Suzuki K, Nakamura K, Mori N. Vldlr overexpression causes hyperactivity in rats. Mol Autism 2012; 3:11. [PMID: 23110844 PMCID: PMC3533969 DOI: 10.1186/2040-2392-3-11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 09/26/2012] [Indexed: 01/05/2023] Open
Abstract
UNLABELLED BACKGROUND Reelin regulates neuronal positioning in cortical brain structures and neuronal migration via binding to the lipoprotein receptors Vldlr and Lrp8. Reeler mutant mice display severe brain morphological defects and behavioral abnormalities. Several reports have implicated reelin signaling in the etiology of neurodevelopmental and psychiatric disorders, including autism, schizophrenia, bipolar disorder, and depression. Moreover, it has been reported that VLDLR mRNA levels are increased in the post-mortem brain of autistic patients. METHODS We generated transgenic (Tg) rats overexpressing Vldlr, and examined their histological and behavioral features. RESULTS Spontaneous locomotor activity was significantly increased in Tg rats, without detectable changes in brain histology. Additionally, Tg rats tended to show performance deficits in the radial maze task, suggesting that their spatial working memory was slightly impaired. Thus, Vldlr levels may be involved in determining locomotor activity and memory function. CONCLUSIONS Unlike reeler mice, patients with neurodevelopmental or psychiatric disorders do not show striking neuroanatomical aberrations. Therefore, it is notable, from a clinical point of view, that we observed behavioral phenotypes in Vldlr-Tg rats in the absence of neuroanatomical abnormalities.
Collapse
Affiliation(s)
- Keiko Iwata
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nobuo Izumo
- Department of Clinical Pharmacology, Yokohama College of Pharmacy, Yokohama, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayuki Manabe
- Division of Gene Expression Mechanism, Institute for Comprehensive Medical Science, Fujita Health University, Toyoake, Japan
| | - Yukiko Ishibashi
- Department of Synthetic Organic Chemistry, Yokohama College of Pharmacy, Yokohama, Japan
| | - Yukio Ichitani
- Institute of Psychology and Behavioral Neuroscience, University of Tsukuba, Tsukuba, Japan
| | - Kazuo Yamada
- Institute of Psychology and Behavioral Neuroscience, University of Tsukuba, Tsukuba, Japan
| | - Ismail Thanseem
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ayyappan Anitha
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mahesh Mundalil Vasu
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chie Shimmura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Taro Takahashi
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yasuhide Iwata
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Katsuaki Suzuki
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazuhiko Nakamura
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Norio Mori
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
31
|
Fatemi SH, Aldinger KA, Ashwood P, Bauman ML, Blaha CD, Blatt GJ, Chauhan A, Chauhan V, Dager SR, Dickson PE, Estes AM, Goldowitz D, Heck DH, Kemper TL, King BH, Martin LA, Millen KJ, Mittleman G, Mosconi MW, Persico AM, Sweeney JA, Webb SJ, Welsh JP. Consensus paper: pathological role of the cerebellum in autism. CEREBELLUM (LONDON, ENGLAND) 2012; 11:777-807. [PMID: 22370873 PMCID: PMC3677555 DOI: 10.1007/s12311-012-0355-9] [Citation(s) in RCA: 464] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
There has been significant advancement in various aspects of scientific knowledge concerning the role of cerebellum in the etiopathogenesis of autism. In the current consensus paper, we will observe the diversity of opinions regarding the involvement of this important site in the pathology of autism. Recent emergent findings in literature related to cerebellar involvement in autism are discussed, including: cerebellar pathology, cerebellar imaging and symptom expression in autism, cerebellar genetics, cerebellar immune function, oxidative stress and mitochondrial dysfunction, GABAergic and glutamatergic systems, cholinergic, dopaminergic, serotonergic, and oxytocin-related changes in autism, motor control and cognitive deficits, cerebellar coordination of movements and cognition, gene-environment interactions, therapeutics in autism, and relevant animal models of autism. Points of consensus include presence of abnormal cerebellar anatomy, abnormal neurotransmitter systems, oxidative stress, cerebellar motor and cognitive deficits, and neuroinflammation in subjects with autism. Undefined areas or areas requiring further investigation include lack of treatment options for core symptoms of autism, vermal hypoplasia, and other vermal abnormalities as a consistent feature of autism, mechanisms underlying cerebellar contributions to cognition, and unknown mechanisms underlying neuroinflammation.
Collapse
Affiliation(s)
- S Hossein Fatemi
- University of Minnesota Medical School, 420 Delaware St. SE, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
BECKER JÜRGEN, FRÖHLICH JOHANNA, PERSKE CHRISTINA, PAVLAKOVIC HELENA, WILTING JÖRG. Reelin signalling in neuroblastoma: Migratory switch in metastatic stages. Int J Oncol 2012; 41:681-9. [DOI: 10.3892/ijo.2012.1488] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/08/2012] [Indexed: 11/05/2022] Open
|
33
|
Hadj-Hamou NS, Laé M, Almeida A, Grange PDL, Kirova Y, Sastre-Garau X, Malfoy B. A transcriptome signature of endothelial lymphatic cells coexists with the chronic oxidative stress signature in radiation-induced post-radiotherapy breast angiosarcomas. Carcinogenesis 2012; 33:1399-405. [DOI: 10.1093/carcin/bgs155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
34
|
Hattori M. [Reelin dysfunction in neuropsychiatric diseases and its potential as a drug target]. Nihon Yakurigaku Zasshi 2012; 139:99-102. [PMID: 22451463 DOI: 10.1254/fpj.139.99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
35
|
Anisuzzaman, Islam MK, Alim MA, Miyoshi T, Hatta T, Yamaji K, Matsumoto Y, Fujisaki K, Tsuji N. Longistatin is an unconventional serine protease and induces protective immunity against tick infestation. Mol Biochem Parasitol 2011; 182:45-53. [PMID: 22206819 DOI: 10.1016/j.molbiopara.2011.12.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 01/28/2023]
Abstract
Classical serine proteases use the conserved Ser/His/Asp catalytic triad to hydrolyze substrates. Here, we show that longistatin, a salivary gland protein with two EF-hand domains from the vector tick Haemaphysalis longicornis, does not have the conserved catalytic triad, but still functions as a serine protease. Longistatin was synthesized in and secreted from the salivary glands of ticks, and is injected into host tissues during the acquisition of blood-meals. Longistatin hydrolyzed fibrinogen, an essential plasma protein in the coagulation cascade, and activated plasminogen, into its active form plasmin, a serine protease that dissolves fibrin clots. Longistatin efficiently hydrolyzed several serine protease-specific substrates showing its specificity to the amide bond of Arg. Longistatin did not hydrolyze synthetic substrates specific for other groups of proteases. The enzyme was active at a wide range of temperatures and pHs, with the optimum at 37°C and pH 7. Its activity was efficiently inhibited by various serine protease inhibitors such as phenylmethanesulfonyl fluoride (PMSF), aprotinin, antipain, and leupeptin with the estimated IC(50) of 278.57 μM, 0.35 μM, 41.56 μM and 198.86 μM, respectively. In addition, longistatin was also potently inhibited by Zinc (Zn(2+)) in a concentration-dependent manner with an IC(50) value of 275 μM, and the inhibitory effect of Zn(2+) was revived by ethylenediaminetetra acetic acid (EDTA). Immunization studies revealed that longistatin sharply induced high levels of protective IgG antibodies against ticks. Immunization with longistatin reduced repletion of ticks by about 54%, post engorgement body weight by >11% and molting of nymphs by approximately 34%; thus, the vaccination trial was approximately 73% effective against tick infestation. Taken together, our results suggest that longistatin is a new potent atypical serine protease, and may be an interesting candidate for the development of anti-tick vaccines.
Collapse
Affiliation(s)
- Anisuzzaman
- Department of Global Agricultural Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Dendritic spines are dynamic structures that accommodate the majority of excitatory synapses in the brain and are influenced by extracellular signals from presynaptic neurons, glial cells, and the extracellular matrix (ECM). The ECM surrounds dendritic spines and extends into the synaptic cleft, maintaining synapse integrity as well as mediating trans-synaptic communications between neurons. Several scaffolding proteins and glycans that compose the ECM form a lattice-like network, which serves as an attractive ground for various secreted glycoproteins, lectins, growth factors, and enzymes. ECM components can control dendritic spines through the interactions with their specific receptors or by influencing the functions of other synaptic proteins. In this review, we focus on ECM components and their receptors that regulate dendritic spine development and plasticity in the normal and diseased brain.
Collapse
Affiliation(s)
- Lorraine E. Dansie
- Division of Biomedical Sciences, Biochemistry and Molecular Biology Program, University of California Riverside, Riverside, California 92521
| | - Iryna M. Ethell
- Division of Biomedical Sciences, Biochemistry and Molecular Biology Program, University of California Riverside, Riverside, California 92521
| |
Collapse
|
37
|
Extracellular proteases in epilepsy. Epilepsy Res 2011; 96:191-206. [DOI: 10.1016/j.eplepsyres.2011.08.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/10/2011] [Accepted: 08/03/2011] [Indexed: 11/20/2022]
|
38
|
Bao YY, Xue J, Wu WJ, Wang Y, Lv ZY, Zhang CX. An immune-induced reeler protein is involved in the Bombyx mori melanization cascade. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2011; 41:696-706. [PMID: 21624461 DOI: 10.1016/j.ibmb.2011.05.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 05/05/2011] [Accepted: 05/12/2011] [Indexed: 05/30/2023]
Abstract
In this study, we isolated two reeler cDNAs from bacteria-challenged larval fat bodies of the silkworm, Bombyx mori. A reeler domain spanned most of the coding regions of these two cDNAs, and their expression patterns were different in B. mori larval tissues. The reeler1 gene was strongly induced by Escherichia coli K12 and Bacillus subtilis in B. mori larval hemocytes, fat bodies and midguts, but reeler2 was expressed at extremely low levels in these tissues. We focused on the reeler1 gene for functional analysis. Interference by double-stranded reeler1 RNA in vivo led to reduced nodule formation in bacteria-injected larvae, while the injection of recombinant Reeler1 promoted nodule formation in reeler1 gene-silenced larvae, indicating that Reeler1 is involved in the nodulation response. Knockdown of the reeler1 gene significantly decreased phenoloxidase activity in bacteria-challenged larval hemolymph, while injection of recombinant Reeler1 enhanced phenoloxidase activity, suggesting that Reeler1 is involved in the prophenoloxidase activation cascade. Our results provide new mechanistic evidence about the melanization cascade in the insect immunity.
Collapse
Affiliation(s)
- Yan-Yuan Bao
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Yuhangtang Road 866, Hangzhou, Zhejiang 310058, China
| | | | | | | | | | | |
Collapse
|
39
|
Wei H, Malik M, Sheikh AM, Merz G, Ted Brown W, Li X. Abnormal cell properties and down-regulated FAK-Src complex signaling in B lymphoblasts of autistic subjects. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:66-74. [PMID: 21703394 DOI: 10.1016/j.ajpath.2011.03.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 03/04/2011] [Accepted: 03/21/2011] [Indexed: 12/26/2022]
Abstract
Recent studies suggest that one of the major pathways to the pathogenesis of autism is reduced cell migration. Focal adhesion kinase (FAK) has an important role in neural migration, dendritic morphological characteristics, axonal branching, and synapse formation. The FAK-Src complex, activated by upstream reelin and integrin β1, can initiate a cascade of phosphorylation events to trigger multiple intracellular pathways, including mitogen-activated protein kinase-extracellular signal-regulated kinase and phosphatidylinositol 3-kinase-Akt signaling. In this study, by using B lymphoblasts as a model, we tested whether integrin β1 and FAK-Src signaling are abnormally regulated in autism and whether abnormal FAK-Src signaling leads to defects in B-lymphoblast adhesion, migration, proliferation, and IgG production. To our knowledge, for the first time, we show that protein expression levels of both integrin β1 and FAK are significantly decreased in autistic lymphoblasts and that Src protein expression and the phosphorylation of an active site (Y416) are also significantly decreased. We also found that lymphoblasts from autistic subjects exhibit significantly decreased migration, increased adhesion properties, and an impaired capacity for IgG production. The overexpression of FAK in autistic lymphoblasts countered the adhesion and migration defects. In addition, we demonstrate that FAK mediates its effect through the activation of Src, phosphatidylinositol 3-kinase-Akt, and mitogen-activated protein kinase signaling cascades and that paxillin is also likely involved in the regulation of adhesion and migration in autistic lymphoblasts.
Collapse
Affiliation(s)
- Hongen Wei
- Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities, New York, New York 10314, USA
| | | | | | | | | | | |
Collapse
|
40
|
Zhao S, Frotscher M. Go or stop? Divergent roles of Reelin in radial neuronal migration. Neuroscientist 2011; 16:421-34. [PMID: 20817919 DOI: 10.1177/1073858410367521] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Neuronal migration is an essential step of brain development and is controlled by a variety of cellular proteins and extracellular matrix molecules. Reelin, an extracellular matrix protein, is required for neuronal migration. Over the past 10 years, the Reelin signaling cascade has been studied intensively. However, the role of Reelin in neuronal migration has remained unclear. Different Reelin fragments and different Reelin receptors suggest multiple functions of Reelin. In this review, the authors focus on Reelin effects on the actin cytoskeleton of migrating neurons.
Collapse
Affiliation(s)
- Shanting Zhao
- Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
41
|
Dutta S, Gangopadhyay PK, Sinha S, Chatterjee A, Ghosh S, Rajamma U. An association analysis of reelin gene (RELN) polymorphisms with childhood epilepsy in eastern Indian population from West Bengal. Cell Mol Neurobiol 2011; 31:45-56. [PMID: 20697953 DOI: 10.1007/s10571-010-9551-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 07/23/2010] [Indexed: 12/28/2022]
Abstract
Epilepsy is a common neurological condition characterized by unprovoked seizure attacks. Early brain developmental abnormalities involving neuronal migration and lamination are implicated in childhood epilepsy. Reelin, a neuronal-signaling molecule plays a crucial role in these migratory processes. Therefore, reelin gene (RELN), which is located on human chromosome 7q22 is considered as a potential candidate gene for childhood epilepsy. In this study, we recruited 63 patients with childhood-onset epilepsy and 103 healthy controls from West Bengal in India. Genomic DNA isolated from leukocytes of cases and control individuals were used for genotyping analysis of 16 markers of RELN. Case-control analysis revealed significant over-representation of G/C and (G/C+C/C) genotypes, and C allele of exon 22 G/C marker (rs362691) in cases as compared to controls. Pair-wise linkage disequilibrium analysis demonstrated two separate LD blocks with moderately high D' values in epileptic cases. Based on these data, we have carried out haplotype case-control analysis. Even though we found over-representation of A-C haplotype of intron 12 A/C/exon 22 G/C markers and haplotype combination involving G-allele of exon 22 marker in cases and controls, respectively, the overall test was not significant. LD in this region involving this marker was also more robust in epileptic cases. Taken together, the results provide possible evidences for association of exon 22 G/C marker or any marker in the vicinity, which is in LD with this marker with epilepsy in the West Bengal population. Further investigations involving higher sample sizes are warranted to validate the present finding.
Collapse
Affiliation(s)
- Shruti Dutta
- Manovikas Biomedical Research and Diagnostic Centre, Manovikas Kendra Rehabilitation and Research Institute for the Handicapped, Kolkata, India
| | | | | | | | | | | |
Collapse
|
42
|
Qin L, Tu W, Sun X, Zhang J, Chen Y, Zhao H. Retardation of neurobehavioral development and reelin down-regulation regulated by further DNA methylation in the hippocampus of the rat pups are associated with maternal deprivation. Behav Brain Res 2010; 217:142-7. [PMID: 20974192 DOI: 10.1016/j.bbr.2010.10.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/12/2010] [Accepted: 10/13/2010] [Indexed: 12/31/2022]
Abstract
It is known that early life stress has profound effects in early developing hippocampus. Reelin is a large protein that regulates neuronal migration during embryonic development. The expression of reelin persists in brain, but its function is little known. The aim of the present study was to investigate the effects of maternal deprivation (MD) on early neurobehavioral development of rats, and the role of reelin and the potential mechanism underlying regulation of its expression in hippocampus. Rat pups were removed from mothers during the postnatal day (PND) 2-15 for 3h a day. Reflex developments including grasping, gait, righting, cliff avoidance, auditory startle, hot-plate test and negative geotaxis, were tested during the first 3 weeks. The level of reelin mRNA and reelin gene methylation in the hippocampal formation were determined using real-time PCR analysis. As expected, some differences appeared in the measure of neurobehavior and expression of reelin in rat pups. Several significant deficiencies were observed in bodyweight, auditory startle and grasping reflex while a great enhancement in hot-plate test in rat pups suffering from MD. On PND 22, the expression of reelin mRNA reduced in the hippocampus followed by MD. Meanwhile, the changes of DNA methylation showed an opposite trend compared with the reelin expression. The results suggest that MD in early life has harmful effects on neurobehavioral development, and causes the down-regulation of reelin mRNA by further DNA methylation in postnatal hippocampus.
Collapse
Affiliation(s)
- Lina Qin
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
Antibodies display great versatility in protein interactions and have become important therapeutic agents for a variety of human diseases. Their ability to discriminate between highly conserved sequences could be of great use for therapeutic approaches that target proteases, for which structural features are conserved among family members. Recent crystal structures of antibody-protease complexes provide exciting insight into the variety of ways antibodies can interfere with the catalytic machinery of serine proteases. The studies revealed the molecular details of two fundamental mechanisms by which antibodies inhibit catalysis of trypsin-like serine proteases, exemplified by hepatocyte growth factor activator and MT-SP1 (matriptase). Enzyme kinetics defines both mechanisms as competitive inhibition systems, yet, on the molecular level, they involve distinct structural elements of the active-site region. In the steric hindrance mechanism, the antibody binds to protruding surface loops and inserts one or two CDR (complementarity-determining region) loops into the enzyme's substrate-binding cleft, which results in obstruction of substrate access. In the allosteric inhibition mechanism the antibody binds outside the active site at the periphery of the substrate-binding cleft and, mediated through a conformational change of a surface loop, imposes structural changes at important substrate interaction sites resulting in impaired catalysis. At the centre of this allosteric mechanism is the 99-loop, which is sandwiched between the substrate and the antibody-binding sites and serves as a mobile conduit between these sites. These findings provide comprehensive structural and functional insight into the molecular versatility of antibodies for interfering with the catalytic machinery of proteases.
Collapse
|
44
|
Cellular form of prion protein inhibits Reelin-mediated shedding of Caspr from the neuronal cell surface to potentiate Caspr-mediated inhibition of neurite outgrowth. J Neurosci 2010; 30:9292-305. [PMID: 20610764 DOI: 10.1523/jneurosci.5657-09.2010] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Extension of axonal and dendritic processes in the CNS is tightly regulated by outgrowth-promoting and -inhibitory cues to assure precision of synaptic connections. We identify a novel role for contactin-associated protein (Caspr) as an inhibitory cue that reduces neurite outgrowth from CNS neurons. We show that proteolysis of Caspr at the cell surface is regulated by the cellular form of prion protein (PrP), which directly binds to Caspr. PrP inhibits Reelin-mediated shedding of Caspr from the cell surface, thereby increasing surface levels of Caspr and potentiating the inhibitory effect of Caspr on neurite outgrowth. PrP deficiency results in reduced levels of Caspr at the cell surface, enhanced neurite outgrowth in vitro, and more efficient regeneration of axons in vivo following spinal cord injury. Thus, we reveal a previously unrecognized role for Caspr and PrP in inhibitory modulation of neurite outgrowth in CNS neurons, which is counterbalanced by the proteolytic activity of Reelin.
Collapse
|
45
|
Khalfallah A, Schrauwen I, Mnaja M, Fransen E, Lahmar I, Ealy M, Dhouib L, Ayadi H, Charfedine I, Driss N, Ghorbel A, Smith RJ, Masmoudi S, Van Camp G. Genetic variants in RELN are associated with otosclerosis in a non-European population from Tunisia. Ann Hum Genet 2010; 74:399-405. [DOI: 10.1111/j.1469-1809.2010.00595.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
46
|
Ealy M, Smith RJ. The Genetics of otosclerosis. Hear Res 2010; 266:70-4. [DOI: 10.1016/j.heares.2009.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 07/03/2009] [Accepted: 07/06/2009] [Indexed: 11/15/2022]
|
47
|
Gundelfinger ED, Frischknecht R, Choquet D, Heine M. Converting juvenile into adult plasticity: a role for the brain’s extracellular matrix. Eur J Neurosci 2010; 31:2156-65. [DOI: 10.1111/j.1460-9568.2010.07253.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
48
|
Abstract
Reelin plays a pivotal role in neurodevelopment. Excessive RELN promoter methylation and/or decreased RELN gene expression have been described in schizophrenia and autism. We assessed RELN promoter methylation in post-mortem temporocortical tissue (Brodmann Area 41/42) of three prepuberal and six postpuberal normal individuals. The former display very little or no methylation, whereas most postpuberal individuals are heavily methylated, especially at CpG positions located between -131 and -98 bp (prepuberal vs. postpuberal, P<0.05). Sex hormones thus seemingly boost DNA methylation at the RELN promoter. This physiological change could significantly contribute to the onset of schizophrenia and the worsening of autistic behaviors, both typically occurring at puberty.
Collapse
|
49
|
Ceruloplasmin-induced aggregation of P19 neurons involves a serine protease activity and is accompanied by reelin cleavage. Neuroscience 2010; 167:633-43. [PMID: 20188154 DOI: 10.1016/j.neuroscience.2010.02.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 02/16/2010] [Accepted: 02/17/2010] [Indexed: 12/27/2022]
Abstract
The cytoarchitectural organization of the nervous system depends partly on extracellular serine proteases, including reelin. This 400K protein, which also exists as the N-terminally-derived 300K and 180K fragments, acts through binding to the lipoprotein receptors apolipoprotein E receptor 2 (ApoER2) and very low-density lipoprotein receptor (VLDLR). Ceruloplasmin (CP), a multifunctional protein found in the circulation and also expressed on glial cells, was shown to bind to, and induce aggregation of neurons newly differentiated from P19 embryonic stem cells. This indicated a potential developmental role of CP in neuronal organization, possibly in relation with reelin and other extracellular serine proteases. Therefore, we analysed the effect of cell-impermeant, large spectrum, serine protease inhibitors on CP-induced neuroaggregation and studied reelin expression. Soybean trypsin inhibitor and aprotinin (SBTI+Apro) inhibited CP neuroaggregative action. Undifferentiated and neurally-differentiating cultures secreted the 400K reelin. The 180K fragment was present during and after differentiation whereas the 300K species was barely detectable. However, CP stimulated generation of the 300K in the differentiated neuronal cultures, and SBTI+Apro abolished this CP effect. Time course profiles and function-blocking antibody indicated that neuroaggregation does not depend on the generation of the 300K fragment or on reelin action. CP neuroaggregative action thus involves a pericellular serine protease, different from reelin. On the other hand, the CP stimulation of reelin cleavage is in line with a possible role of CP in nervous system development. Since P19 cells express ApoER2 and VLDLR, they can help understanding relationships existing between CP, reelin and intervening protease(s).
Collapse
|
50
|
Tseng WL, Huang CL, Chong KY, Liao CH, Stern A, Cheng JC, Tseng CP. Reelin is a platelet protein and functions as a positive regulator of platelet spreading on fibrinogen. Cell Mol Life Sci 2010; 67:641-53. [PMID: 19936619 PMCID: PMC11115871 DOI: 10.1007/s00018-009-0201-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 10/06/2009] [Accepted: 11/02/2009] [Indexed: 12/21/2022]
Abstract
Abnormalities of platelet functions have been linked to reelin-impaired neuronal disorders. However, little attention has been given to understanding the interplay between reelin and platelet. In this study, reelin was found to present in the human platelets and megakaryocyte-like leukemic cells. Reelin-binding assays revealed that extracellular reelin can interact with platelets through the receptor belonging to the low density lipoprotein receptor gene family. The reelin-to-platelet interactions enhance platelet spreading on fibrinogen concomitant with the augmentation of lamellipodia formation and F-actin bundling. In contrast, reelin has no effect on integrin alphaIIbbeta3 activation and agonist-induced platelet aggregation. Molecular analysis revealed that the up-regulation of Rac1 activity and the inhibition of protein kinase C delta-Thr505 phosphorylation are important for reelin-mediated enhancement of platelet spreading on fibrinogen. These findings demonstrate for the first time that reelin is present in platelets and the reelin-to-platelet interactions play a novel role in platelet signaling and functions.
Collapse
Affiliation(s)
- Wei-Lien Tseng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kweishan, Taoyuan, 333 Taiwan, ROC
| | - Chien-Ling Huang
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333 Taiwan, ROC
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333 Taiwan, ROC
| | - Chang-Huei Liao
- Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333 Taiwan, ROC
| | - Arnold Stern
- Department of Pharmacology, New York University School of Medicine, New York, NY USA
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404 Taiwan, ROC
| | - Ching-Ping Tseng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kweishan, Taoyuan, 333 Taiwan, ROC
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, 259 Wen-Hwa 1st Road, Kweishan, Taoyuan, 333 Taiwan, ROC
| |
Collapse
|