1
|
Sah S, Keable R, Pfundstein G, Clemens KJ, Begg D, Schachner M, Leshchyns'ka I, Sytnyk V. Deficiency in the neural cell adhesion molecule 2 (NCAM2) reduces axonal levels of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), affects axonal organization in the hippocampus, and leads to behavioral deficits. Cereb Cortex 2023; 33:10047-10065. [PMID: 37522285 DOI: 10.1093/cercor/bhad264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/25/2023] [Accepted: 01/26/2023] [Indexed: 08/01/2023] Open
Abstract
The neural cell adhesion molecule 2 (NCAM2) regulates axonal organization in the central nervous system via mechanisms that have remained poorly understood. We now show that NCAM2 increases axonal levels of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), a protease that regulates axonal guidance. In brains of NCAM2-deficient mice, BACE1 levels are reduced in hippocampal mossy fiber projections, and the infrapyramidal bundle of these projections is shortened. This abnormal axonal organization correlates with impaired short-term spatial memory and cognitive flexibility in NCAM2-deficient male and female mice. Self-grooming, rearing, digging and olfactory acuity are increased in NCAM2-deficient male mice, when compared with littermate wild-type mice of the same sex. NCAM2-deficient female mice also show increased self-grooming, but are reduced in rearing, and do not differ from female wild-type mice in olfactory acuity and digging behavior. Our results indicate that errors in axonal guidance and organization caused by impaired BACE1 function can underlie the manifestation of neurodevelopmental disorders, including autism as found in humans with deletions of the NCAM2 gene.
Collapse
Affiliation(s)
- Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Kelly J Clemens
- School of Psychology, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Denovan Begg
- School of Psychology, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, United States
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
2
|
Moore A, Chinnaiya K, Kim DW, Brown S, Stewart I, Robins S, Dowsett GKC, Muir C, Travaglio M, Lewis JE, Ebling F, Blackshaw S, Furley A, Placzek M. Loss of Function of the Neural Cell Adhesion Molecule NrCAM Regulates Differentiation, Proliferation and Neurogenesis in Early Postnatal Hypothalamic Tanycytes. Front Neurosci 2022; 16:832961. [PMID: 35464310 PMCID: PMC9022636 DOI: 10.3389/fnins.2022.832961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Hypothalamic tanycytes are neural stem and progenitor cells, but little is known of how they are regulated. Here we provide evidence that the cell adhesion molecule, NrCAM, regulates tanycytes in the adult niche. NrCAM is strongly expressed in adult mouse tanycytes. Immunohistochemical and in situ hybridization analysis revealed that NrCAM loss of function leads to both a reduced number of tanycytes and reduced expression of tanycyte-specific cell markers, along with a small reduction in tyrosine hydroxylase-positive arcuate neurons. Similar analyses of NrCAM mutants at E16 identify few changes in gene expression or cell composition, indicating that NrCAM regulates tanycytes, rather than early embryonic hypothalamic development. Neurosphere and organotypic assays support the idea that NrCAM governs cellular homeostasis. Single-cell RNA sequencing (scRNA-Seq) shows that tanycyte-specific genes, including a number that are implicated in thyroid hormone metabolism, show reduced expression in the mutant mouse. However, the mild tanycyte depletion and loss of markers observed in NrCAM-deficient mice were associated with only a subtle metabolic phenotype.
Collapse
Affiliation(s)
- Alex Moore
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Kavitha Chinnaiya
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah Brown
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Iain Stewart
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Sarah Robins
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Georgina K. C. Dowsett
- Wellcome Trust-Medical Research Council Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte Muir
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Marco Travaglio
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jo E. Lewis
- Wellcome Trust-Medical Research Council Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Fran Ebling
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrew Furley
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Marysia Placzek
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
3
|
Oleari R, Massa V, Cariboni A, Lettieri A. The Differential Roles for Neurodevelopmental and Neuroendocrine Genes in Shaping GnRH Neuron Physiology and Deficiency. Int J Mol Sci 2021; 22:9425. [PMID: 34502334 PMCID: PMC8431607 DOI: 10.3390/ijms22179425] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 01/19/2023] Open
Abstract
Gonadotropin releasing hormone (GnRH) neurons are hypothalamic neuroendocrine cells that control sexual reproduction. During embryonic development, GnRH neurons migrate from the nose to the hypothalamus, where they receive inputs from several afferent neurons, following the axonal scaffold patterned by nasal nerves. Each step of GnRH neuron development depends on the orchestrated action of several molecules exerting specific biological functions. Mutations in genes encoding for these essential molecules may cause Congenital Hypogonadotropic Hypogonadism (CHH), a rare disorder characterized by GnRH deficiency, delayed puberty and infertility. Depending on their action in the GnRH neuronal system, CHH causative genes can be divided into neurodevelopmental and neuroendocrine genes. The CHH genetic complexity, combined with multiple inheritance patterns, results in an extreme phenotypic variability of CHH patients. In this review, we aim at providing a comprehensive and updated description of the genes thus far associated with CHH, by dissecting their biological relevance in the GnRH system and their functional relevance underlying CHH pathogenesis.
Collapse
Affiliation(s)
- Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy;
| | - Valentina Massa
- Department of Health Sciences, University of Milan, 20142 Milano, Italy;
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milano, Italy
| | - Anna Cariboni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy;
| | - Antonella Lettieri
- Department of Health Sciences, University of Milan, 20142 Milano, Italy;
- CRC Aldo Ravelli for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, 20142 Milano, Italy
| |
Collapse
|
4
|
Chen Y, Sun T, Niu Y, Wang D, Liu K, Wang T, Wang S, Xu H, Liu J. Cell adhesion molecule L1 like plays a role in the pathogenesis of idiopathic hypogonadotropic hypogonadism. J Endocrinol Invest 2021; 44:1739-1751. [PMID: 33453020 DOI: 10.1007/s40618-020-01485-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE The pathogenesis of idiopathic hypogonadotropic hypogonadism (IHH) is genetically complex. The aims of this study were to investigate the genetic profile and clinical manifestation of IHH in a Chinese pedigree and to discover new IHH-associated genes. METHODS The first step was to follow up the clinical phenotype and therapeutic outcomes of the pedigree in university hospital. The second step was that mutation screening was performed in this pedigree and 100 healthy controls. The third step was to further verify the pathogenicity of the discovered rare sequencing variant (RSV) by functional experiments. Whole exome sequencing, Sanger sequencing, testicular volume (TV), semen analysis, assessment of cell migration and necroptosis were performed. RESULTS One heterozygous RSV (p.G517E) in CHL1 was identified in two male IHH patients and their mother in the pedigree, but not in healthy controls. All the three individuals exhibited olfactory impairment. hCG/hMG treatment significantly improved TV, serum testosterone and/or semen parameters of the two male patients. Functional analysis indicated that CHL1 significantly regulated GnRH neuronal cell line (GN11 cells) migration and necroptosis, with alteration of ERK1/2 activation, calcium loading, and transcription of RIPK3 and MLKL. However, the above processes were negatively influenced by the CHL1 RSV. CONCLUSIONS Our study reports the genetic relevance of CHL1 in IHH, and characterizes the phenotypic and therapeutic profiles in patients carrying the CHL1 RSV. CHL1 may act as a new IHH-associated gene, and should be taken into consideration in future investigations for this field.
Collapse
Affiliation(s)
- Y Chen
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - T Sun
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Y Niu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - D Wang
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - K Liu
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - T Wang
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - S Wang
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - H Xu
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - J Liu
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Department of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
5
|
Cntn4, a risk gene for neuropsychiatric disorders, modulates hippocampal synaptic plasticity and behavior. Transl Psychiatry 2021; 11:106. [PMID: 33542194 PMCID: PMC7862349 DOI: 10.1038/s41398-021-01223-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/05/2021] [Accepted: 01/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neurodevelopmental and neuropsychiatric disorders, such as autism spectrum disorders (ASD), anorexia nervosa (AN), Alzheimer's disease (AD), and schizophrenia (SZ), are heterogeneous brain disorders with unknown etiology. Genome wide studies have revealed a wide variety of risk genes for these disorders, indicating a biological link between genetic signaling pathways and brain pathology. A unique risk gene is Contactin 4 (Cntn4), an Ig cell adhesion molecule (IgCAM) gene, which has been associated with several neuropsychiatric disorders including ASD, AN, AD, and SZ. Here, we investigated the Cntn4 gene knockout (KO) mouse model to determine whether memory dysfunction and altered brain plasticity, common neuropsychiatric symptoms, are affected by Cntn4 genetic disruption. For that purpose, we tested if Cntn4 genetic disruption affects CA1 synaptic transmission and the ability to induce LTP in hippocampal slices. Stimulation in CA1 striatum radiatum significantly decreased synaptic potentiation in slices of Cntn4 KO mice. Neuroanatomical analyses showed abnormal dendritic arborization and spines of hippocampal CA1 neurons. Short- and long-term recognition memory, spatial memory, and fear conditioning responses were also assessed. These behavioral studies showed increased contextual fear conditioning in heterozygous and homozygous KO mice, quantified by a gene-dose dependent increase in freezing response. In comparison to wild-type mice, Cntn4-deficient animals froze significantly longer and groomed more, indicative of increased stress responsiveness under these test conditions. Our electrophysiological, neuro-anatomical, and behavioral results in Cntn4 KO mice suggest that Cntn4 has important functions related to fear memory possibly in association with the neuronal morphological and synaptic plasticity changes in hippocampus CA1 neurons.
Collapse
|
6
|
Spatiotemporal processing of neural cell adhesion molecules 1 and 2 by BACE1 in vivo. J Biol Chem 2021; 296:100372. [PMID: 33548223 PMCID: PMC7949136 DOI: 10.1016/j.jbc.2021.100372] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Neural cell adhesion molecules 1 (NCAM1) and 2 (NCAM2) belong to the cell adhesion molecules of the immunoglobulin superfamily and have been shown to regulate formation, maturation, and maintenance of synapses. NCAM1 and NCAM2 undergo proteolysis, but the identity of all the proteases involved and how proteolysis is used to regulate their functions are not known. We report here that NCAM1 and NCAM2 are BACE1 substrates in vivo. NCAM1 and NCAM2 overexpressed in HEK cells were both cleaved by metalloproteinases or BACE1, and NCAM2 was also processed by γ-secretase. We identified the BACE1 cleavage site of NCAM1 (at Glu 671) and NCAM2 (at Glu 663) using mass spectrometry and site-directed mutagenesis. Next, we assessed BACE1-mediated processing of NCAM1 and NCAM2 in the mouse brain during aging. NCAM1 and NCAM2 were cleaved in the olfactory bulb of BACE1+/+ but not BACE1−/− mice at postnatal day 10 (P10), 4 and 12 months of age. In the hippocampus, a BACE1-specific soluble fragment of NCAM1 (sNCAM1β) was only detected at P10. However, we observed an accumulation of full-length NCAM1 in hippocampal synaptosomes in 4-month-old BACE1−/− mice. We also found that polysialylated NCAM1 (PSA-NCAM1) levels were increased in BACE1−/− mice at P10 and demonstrated that BACE1 cleaves both NCAM1 and PSA-NCAM1 in vitro. In contrast, we did not find evidence for BACE1-dependent NCAM2 processing in the hippocampus at any age analyzed. In summary, our data demonstrate that BACE1 differentially processes NCAM1 and NCAM2 depending on the region of brain, subcellular localization, and age in vivo.
Collapse
|
7
|
Gandawijaya J, Bamford RA, Burbach JPH, Oguro-Ando A. Cell Adhesion Molecules Involved in Neurodevelopmental Pathways Implicated in 3p-Deletion Syndrome and Autism Spectrum Disorder. Front Cell Neurosci 2021; 14:611379. [PMID: 33519384 PMCID: PMC7838543 DOI: 10.3389/fncel.2020.611379] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/15/2020] [Indexed: 01/06/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterized by impaired social interaction, language delay and repetitive or restrictive behaviors. With increasing prevalence, ASD is currently estimated to affect 0.5–2.0% of the global population. However, its etiology remains unclear due to high genetic and phenotypic heterogeneity. Copy number variations (CNVs) are implicated in several forms of syndromic ASD and have been demonstrated to contribute toward ASD development by altering gene dosage and expression. Increasing evidence points toward the p-arm of chromosome 3 (chromosome 3p) as an ASD risk locus. Deletions occurring at chromosome 3p result in 3p-deletion syndrome (Del3p), a rare genetic disorder characterized by developmental delay, intellectual disability, facial dysmorphisms and often, ASD or ASD-associated behaviors. Therefore, we hypothesize that overlapping molecular mechanisms underlie the pathogenesis of Del3p and ASD. To investigate which genes encoded in chromosome 3p could contribute toward Del3p and ASD, we performed a comprehensive literature review and collated reports investigating the phenotypes of individuals with chromosome 3p CNVs. We observe that high frequencies of CNVs occur in the 3p26.3 region, the terminal cytoband of chromosome 3p. This suggests that CNVs disrupting genes encoded within the 3p26.3 region are likely to contribute toward the neurodevelopmental phenotypes observed in individuals affected by Del3p. The 3p26.3 region contains three consecutive genes encoding closely related neuronal immunoglobulin cell adhesion molecules (IgCAMs): Close Homolog of L1 (CHL1), Contactin-6 (CNTN6), and Contactin-4 (CNTN4). CNVs disrupting these neuronal IgCAMs may contribute toward ASD phenotypes as they have been associated with key roles in neurodevelopment. CHL1, CNTN6, and CNTN4 have been observed to promote neurogenesis and neuronal survival, and regulate neuritogenesis and synaptic function. Furthermore, there is evidence that these neuronal IgCAMs possess overlapping interactomes and participate in common signaling pathways regulating axon guidance. Notably, mouse models deficient for these neuronal IgCAMs do not display strong deficits in axonal migration or behavioral phenotypes, which is in contrast to the pronounced defects in neuritogenesis and axon guidance observed in vitro. This suggests that when CHL1, CNTN6, or CNTN4 function is disrupted by CNVs, other neuronal IgCAMs may suppress behavioral phenotypes by compensating for the loss of function.
Collapse
Affiliation(s)
- Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Rosemary A Bamford
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - J Peter H Burbach
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
8
|
Zheng Y, Verhoeff TA, Perez Pardo P, Garssen J, Kraneveld AD. The Gut-Brain Axis in Autism Spectrum Disorder: A Focus on the Metalloproteases ADAM10 and ADAM17. Int J Mol Sci 2020; 22:ijms22010118. [PMID: 33374371 PMCID: PMC7796333 DOI: 10.3390/ijms22010118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is a spectrum of disorders that are characterized by problems in social interaction and repetitive behavior. The disease is thought to develop from changes in brain development at an early age, although the exact mechanisms are not known yet. In addition, a significant number of people with ASD develop problems in the intestinal tract. A Disintegrin And Metalloproteases (ADAMs) include a group of enzymes that are able to cleave membrane-bound proteins. ADAM10 and ADAM17 are two members of this family that are able to cleave protein substrates involved in ASD pathogenesis, such as specific proteins important for synapse formation, axon signaling and neuroinflammation. All these pathological mechanisms are involved in ASD. Besides the brain, ADAM10 and ADAM17 are also highly expressed in the intestines. ADAM10 and ADAM17 have implications in pathways that regulate gut permeability, homeostasis and inflammation. These metalloproteases might be involved in microbiota-gut-brain axis interactions in ASD through the regulation of immune and inflammatory responses in the intestinal tract. In this review, the potential roles of ADAM10 and ADAM17 in the pathology of ASD and as targets for new therapies will be discussed, with a focus on the gut-brain axis.
Collapse
Affiliation(s)
- Yuanpeng Zheng
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Tessa A. Verhoeff
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Global Centre of Excellence Immunology, Danone Nutricia Research B.V., 3584CT Utrecht, The Netherlands
| | - Aletta D. Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584CG Utrecht, The Netherlands; (Y.Z.); (T.A.V.); (P.P.P.); (J.G.)
- Correspondence: ; Tel.: +31-(0)3-02534509
| |
Collapse
|
9
|
Brummer T, Müller SA, Pan-Montojo F, Yoshida F, Fellgiebel A, Tomita T, Endres K, Lichtenthaler SF. NrCAM is a marker for substrate-selective activation of ADAM10 in Alzheimer's disease. EMBO Mol Med 2020; 11:emmm.201809695. [PMID: 30833305 PMCID: PMC6460357 DOI: 10.15252/emmm.201809695] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The metalloprotease ADAM10 is a drug target in Alzheimer's disease, where it cleaves the amyloid precursor protein (APP) and lowers amyloid‐beta. Yet, ADAM10 has additional substrates, which may cause mechanism‐based side effects upon therapeutic ADAM10 activation. However, they may also serve—in addition to APP—as biomarkers to monitor ADAM10 activity in patients and to develop APP‐selective ADAM10 activators. Our study demonstrates that one such substrate is the neuronal cell adhesion protein NrCAM. ADAM10 controlled NrCAM surface levels and regulated neurite outgrowth in vitro in an NrCAM‐dependent manner. However, ADAM10 cleavage of NrCAM, in contrast to APP, was not stimulated by the ADAM10 activator acitretin, suggesting that substrate‐selective ADAM10 activation may be feasible. Indeed, a whole proteome analysis of human CSF from a phase II clinical trial showed that acitretin, which enhanced APP cleavage by ADAM10, spared most other ADAM10 substrates in brain, including NrCAM. Taken together, this study demonstrates an NrCAM‐dependent function for ADAM10 in neurite outgrowth and reveals that a substrate‐selective, therapeutic ADAM10 activation is possible and may be monitored with NrCAM.
Collapse
Affiliation(s)
- Tobias Brummer
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stephan A Müller
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany
| | - Francisco Pan-Montojo
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Department of Neurology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Fumiaki Yoshida
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Andreas Fellgiebel
- Department of Psychiatry and Psychotherapy, University Medical Center JGU, Mainz, Germany
| | - Taisuke Tomita
- Laboratory of Neuropathology and Neuroscience, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center JGU, Mainz, Germany
| | - Stefan F Lichtenthaler
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany .,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute for Advanced Study, Technische Universität München, Garching, Germany
| |
Collapse
|
10
|
Bhandari R, Paliwal JK, Kuhad A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. ADVANCES IN NEUROBIOLOGY 2020; 24:97-141. [PMID: 32006358 DOI: 10.1007/978-3-030-30402-7_4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Autism spectrum disorder (ASD) is a complex heterogeneous consortium of pervasive development disorders (PDD) which ranges from atypical autism, autism, and Asperger syndrome affecting brain in the developmental stage. This debilitating neurodevelopmental disorder results in both core as well as associated symptoms. Core symptoms observed in autistic patients are lack of social interaction, pervasive, stereotyped, and restricted behavior while the associated symptoms include irritability, anxiety, aggression, and several comorbid disorders.ASD is a polygenic disorder and is multifactorial in origin. Copy number variations (CNVs) of several genes that regulate the synaptogenesis and signaling pathways are one of the major factors responsible for the pathogenesis of autism. The complex integration of various CNVs cause mutations in the genes which code for molecules involved in cell adhesion, voltage-gated ion-channels, scaffolding proteins as well as signaling pathways (PTEN and mTOR pathways). These mutated genes are responsible for affecting synaptic transmission by causing plasticity dysfunction responsible, in turn, for the expression of ASD.Epigenetic modifications affecting DNA transcription and various pre-natal and post-natal exposure to a variety of environmental factors are also precipitating factors for the occurrence of ASD. All of these together cause dysregulation of glutamatergic signaling as well as imbalance in excitatory: inhibitory pathways resulting in glial cell activation and release of inflammatory mediators responsible for the aberrant social behavior which is observed in autistic patients.In this chapter we review and provide insight into the intricate integration of various genetic, epigenetic, and environmental factors which play a major role in the pathogenesis of this disorder and the mechanistic approach behind this integration.
Collapse
Affiliation(s)
- Ranjana Bhandari
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Jyoti K Paliwal
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India
| | - Anurag Kuhad
- Pharmacology Research Laboratory, University Institute of Pharmaceutical Sciences, UGC-Centre of Advanced Study, Panjab University, Chandigarh, India.
| |
Collapse
|
11
|
Hsia HE, Tüshaus J, Brummer T, Zheng Y, Scilabra SD, Lichtenthaler SF. Functions of 'A disintegrin and metalloproteases (ADAMs)' in the mammalian nervous system. Cell Mol Life Sci 2019; 76:3055-3081. [PMID: 31236626 PMCID: PMC11105368 DOI: 10.1007/s00018-019-03173-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/31/2022]
Abstract
'A disintegrin and metalloproteases' (ADAMs) are a family of transmembrane proteins with diverse functions in multicellular organisms. About half of the ADAMs are active metalloproteases and cleave numerous cell surface proteins, including growth factors, receptors, cytokines and cell adhesion proteins. The other ADAMs have no catalytic activity and function as adhesion proteins or receptors. Some ADAMs are ubiquitously expressed, others are expressed tissue specifically. This review highlights functions of ADAMs in the mammalian nervous system, including their links to diseases. The non-proteolytic ADAM11, ADAM22 and ADAM23 have key functions in neural development, myelination and synaptic transmission and are linked to epilepsy. Among the proteolytic ADAMs, ADAM10 is the best characterized one due to its substrates Notch and amyloid precursor protein, where cleavage is required for nervous system development or linked to Alzheimer's disease (AD), respectively. Recent work demonstrates that ADAM10 has additional substrates and functions in the nervous system and its substrate selectivity may be regulated by tetraspanins. New roles for other proteolytic ADAMs in the nervous system are also emerging. For example, ADAM8 and ADAM17 are involved in neuroinflammation. ADAM17 additionally regulates neurite outgrowth and myelination and its activity is controlled by iRhoms. ADAM19 and ADAM21 function in regenerative processes upon neuronal injury. Several ADAMs, including ADAM9, ADAM10, ADAM15 and ADAM30, are potential drug targets for AD. Taken together, this review summarizes recent progress concerning substrates and functions of ADAMs in the nervous system and their use as drug targets for neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Hung-En Hsia
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany
| | - Tobias Brummer
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany
| | - Yuanpeng Zheng
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany
| | - Simone D Scilabra
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany
- Fondazione Ri.MED, Department of Research, IRCCS-ISMETT, via Tricomi 5, 90127, Palermo, Italy
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany.
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Science, Technische Universität München, 81675, Munich, Germany.
- Munich Center for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
12
|
Ou-Yang MH, Kurz JE, Nomura T, Popovic J, Rajapaksha TW, Dong H, Contractor A, Chetkovich DM, Tourtellotte WG, Vassar R. Axonal organization defects in the hippocampus of adult conditional BACE1 knockout mice. Sci Transl Med 2018; 10:eaao5620. [PMID: 30232227 PMCID: PMC11017370 DOI: 10.1126/scitranslmed.aao5620] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 05/21/2018] [Indexed: 12/14/2022]
Abstract
β-Site APP (amyloid precursor protein) cleaving enzyme 1 (BACE1) is the β-secretase enzyme that initiates production of the toxic amyloid-β peptide that accumulates in the brains of patients with Alzheimer's disease (AD). Hence, BACE1 is a prime therapeutic target, and several BACE1 inhibitor drugs are currently being tested in clinical trials for AD. However, the safety of BACE1 inhibition is unclear. Germline BACE1 knockout mice have multiple neurological phenotypes, although these could arise from BACE1 deficiency during development. To address this question, we report that tamoxifen-inducible conditional BACE1 knockout mice in which the Bace1 gene was ablated in the adult largely lacked the phenotypes observed in germline BACE1 knockout mice. However, one BACE1-null phenotype was induced after Bace1 gene deletion in the adult mouse brain. This phenotype showed reduced length and disorganization of the hippocampal mossy fiber infrapyramidal bundle, the axonal pathway of dentate gyrus granule cells that is maintained by neurogenesis in the mouse brain. This defect in axonal organization correlated with reduced BACE1-mediated cleavage of the neural cell adhesion protein close homolog of L1 (CHL1), which has previously been associated with axon guidance. Although our results indicate that BACE1 inhibition in the adult mouse brain may avoid phenotypes associated with BACE1 deficiency during embryonic and postnatal development, they also suggest that BACE1 inhibitor drugs developed for treating AD may disrupt the organization of an axonal pathway in the hippocampus, an important structure for learning and memory.
Collapse
Affiliation(s)
- Ming-Hsuan Ou-Yang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jonathan E Kurz
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Toshihiro Nomura
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Jelena Popovic
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tharinda W Rajapaksha
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Anis Contractor
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Dane M Chetkovich
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Warren G Tourtellotte
- Department of Neurology and Clinical Neurosciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Division of Neuropathology, Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Robert Vassar
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
13
|
Harley RJ, Murdy JP, Wang Z, Kelly MC, Ropp TJF, Park SH, Maness PF, Manis PB, Coate TM. Neuronal cell adhesion molecule (NrCAM) is expressed by sensory cells in the cochlea and is necessary for proper cochlear innervation and sensory domain patterning during development. Dev Dyn 2018; 247:934-950. [PMID: 29536590 PMCID: PMC6105381 DOI: 10.1002/dvdy.24629] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 03/06/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND In the cochlea, auditory development depends on precise patterns of innervation by afferent and efferent nerve fibers, as well as a stereotyped arrangement of hair and supporting cells. Neuronal cell adhesion molecule (NrCAM) is a homophilic cell adhesion molecule that controls diverse aspects of nervous system development, but the function of NrCAM in cochlear development is not well understood. RESULTS Throughout cochlear innervation, NrCAM is detectable on spiral ganglion neuron (SGN) afferent and olivocochlear efferent fibers, and on the membranes of developing hair and supporting cells. Neonatal Nrcam-null cochleae show errors in type II SGN fasciculation, reduced efferent innervation, and defects in the stereotyped packing of hair and supporting cells. Nrcam loss also leads to dramatic changes in the profiles of presynaptic afferent and efferent synaptic markers at the time of hearing onset. Despite these numerous developmental defects, Nrcam-null adults do not show defects in auditory acuity, and by postnatal day 21, the developmental deficits in ribbon synapse distribution and sensory domain structure appear to have been corrected. CONCLUSIONS NrCAM is expressed by several neural and sensory epithelial subtypes within the developing cochlea, and the loss of Nrcam confers numerous, but nonpermanent, developmental defects in innervation and sensory domain patterning. Developmental Dynamics 247:934-950, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Randall J. Harley
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Joseph P. Murdy
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Zhirong Wang
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Michael C. Kelly
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 35 Convent Dr., Bethesda, MD 20892, USA
| | - Tessa-Jonne F. Ropp
- Department of Otolaryngology/Head and Neck Surgery, The University of North Carolina at Chapel Hill, B251 Marsico Hall, CB#7070, 125 Mason Farm Rd., Chapel Hill, NC 27599, USA
| | - SeHoon H. Park
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, The University of North Carolina School of Medicine, 120 Mason Farm Rd., suite 3020, CB#7260, Chapel Hill, NC 27599, USA
| | - Paul B. Manis
- Department of Otolaryngology/Head and Neck Surgery and Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, B027 Marsico Hall, CB#7070. 125 Mason Farm Rd., Chapel Hill, NC 27599
| | - Thomas M. Coate
- Department of Biology, Georgetown University, 37 and O St. NW, Regents Hall 410, Washington, DC 20007, USA
| |
Collapse
|
14
|
Mu D, Xu Y, Zhao T, Watanabe K, Xiao Z, Ye H. Cntn6 deficiency impairs allocentric navigation in mice. Brain Behav 2018; 8:e00969. [PMID: 30106251 PMCID: PMC5991572 DOI: 10.1002/brb3.969] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION CNTN6 is an immunoglobulin domain-containing cell adhesion molecule that belongs to the contactin family. It is involved in the development of the nervous system. We aim to determine the effect of Cntn6 deficiency on the allocentric navigation in mice. METHODS We recorded the travel distance and escape time of wild-type and Cntn6 mutant male and female mice in the Morris water maze task according to the protocol. RESULTS There was hardly any Cntn6 expression in the hippocampus of postnatal day 0 (P0) mice, while obvious Cntn6 expression was present in the hippocampal CA1 region of the P7 mice. During the acquisition period of Morris water maze task (Day 1 to 4), Cntn6-/- male mice failed to shorten the escape time to reach platform on the third day, while the travel distance to platform was not significantly different. There was no significant difference in both escape time and travel distance to the platform among all female subjects. In the probe trial test (Day 5), spatial memory of the female mutant mice was mildly affected, while Cntn6-/- male mice were normal. In the spatial relearning test (Day 7 to 10), Cntn6-/- male mice showed no difference in escape time to the platform compared to the wild-type male mice, while Cntn6 deficient female mice required shorter escape time to travel to the platform on day 7, day 8, and day 10. CONCLUSIONS Cntn6 is expressed in the developing hippocampus in mice. Cntn6 deficiency affects spatial learning and memory, indicating that Cntn6 plays a role in the development of hippocampus and affects allocentric navigation of the animals.
Collapse
Affiliation(s)
- Di Mu
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Yiliang Xu
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Tian Zhao
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| | - Kazutada Watanabe
- Department of BioengineeringNagaoka University of TechnologyNagaokaNiigataJapan
| | - Zhi‐Cheng Xiao
- The Key Laboratory of Stem Cell and Regenerative MedicineInstitute of Molecular and Clinical MedicineKunming Medical UniversityKunmingChina
- Department of Anatomy and Developmental BiologyMonash UniversityClaytonMELAustralia
| | - Haihong Ye
- Department of Medical Genetics and Developmental BiologySchool of Basic Medical SciencesBeijing Institute for Brain DisordersCenter of SchizophreniaCapital Medical UniversityBeijingChina
| |
Collapse
|
15
|
Kim W, Ma L, Lomoio S, Willen R, Lombardo S, Dong J, Haydon PG, Tesco G. BACE1 elevation engendered by GGA3 deletion increases β-amyloid pathology in association with APP elevation and decreased CHL1 processing in 5XFAD mice. Mol Neurodegener 2018; 13:6. [PMID: 29391027 PMCID: PMC5796504 DOI: 10.1186/s13024-018-0239-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/24/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND β-site amyloid precursor protein cleaving enzyme 1 (BACE1) is the rate-limiting enzyme in the production of amyloid beta (Aβ), the toxic peptide that accumulates in the brains of Alzheimer's disease (AD) patients. Our previous studies have shown that the clathrin adaptor Golgi-localized γ-ear-containing ARF binding protein 3 (GGA3) plays a key role in the trafficking of BACE1 to lysosomes, where it is normally degraded. GGA3 depletion results in BACE1 stabilization both in vitro and in vivo. Moreover, levels of GGA3 are reduced and inversely related to BACE1 levels in post-mortem brains of AD patients. METHOD In order to assess the effect of GGA3 deletion on AD-like phenotypes, we crossed GGA3 -/- mice with 5XFAD mice. BACE1-mediated processing of APP and the cell adhesion molecule L1 like protein (CHL1) was measured as well as levels of Aβ42 and amyloid burden. RESULTS In 5XFAD mice, we found that hippocampal and cortical levels of GGA3 decreased while BACE1 levels increased with age, similar to what is observed in human AD brains. GGA3 deletion prevented age-dependent elevation of BACE1 in GGA3KO;5XFAD mice. We also found that GGA3 deletion resulted in increased hippocampal levels of Aβ42 and amyloid burden in 5XFAD mice at 12 months of age. While levels of BACE1 did not change with age and gender in GGAKO;5XFAD mice, amyloid precursor protein (APP) levels increased with age and were higher in female mice. Moreover, elevation of APP was associated with a decreased BACE1-mediated processing of CHL1 not only in 12 months old 5XFAD mice but also in human brains from subjects affected by Down syndrome, most likely due to substrate competition. CONCLUSION This study demonstrates that GGA3 depletion is a leading candidate mechanism underlying elevation of BACE1 in AD. Furthermore, our findings suggest that BACE1 inhibition could exacerbate mechanism-based side effects in conditions associated with APP elevation (e.g. Down syndrome) owing to impairment of BACE1-mediated processing of CHL1. Therefore, therapeutic approaches aimed to restore GGA3 function and to prevent the down stream effects of its depletion (e.g. BACE1 elevation) represent an attractive alternative to BACE inhibition for the prevention/treatment of AD.
Collapse
Affiliation(s)
- WonHee Kim
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Liang Ma
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Selene Lomoio
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Rachel Willen
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Sylvia Lombardo
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Jinghui Dong
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Philip G. Haydon
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| | - Giuseppina Tesco
- Alzheimer’s Disease Research Laboratory, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111 USA
| |
Collapse
|
16
|
Kleijer KTE, van Nieuwenhuize D, Spierenburg HA, Gregorio-Jordan S, Kas MJH, Burbach JPH. Structural abnormalities in the primary somatosensory cortex and a normal behavioral profile in Contactin-5 deficient mice. Cell Adh Migr 2017; 12:5-18. [PMID: 28346043 PMCID: PMC5810773 DOI: 10.1080/19336918.2017.1288788] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Contactin-5 (Cntn5) is an immunoglobulin cell adhesion molecule that is exclusively expressed in the central nervous system. In view of its association with neurodevelopmental disorders, particularly autism spectrum disorder (ASD), this study focused on Cntn5-positive areas in the forebrain and aimed to explore the morphological and behavioral phenotypes of the Cntn5 null mutant (Cntn5−/−) mouse in relation to these areas and ASD symptomatology. A newly generated antibody enabled us to elaborately describe the spatial expression pattern of Cntn5 in P7 wild type (Cntn5+/+) mice. The Cntn5 expression pattern included strong expression in the cerebral cortex, hippocampus and mammillary bodies in addition to described previously brain nuclei of the auditory pathway and the dorsal thalamus. Thinning of the primary somatosensory (S1) cortex was found in Cntn5−/− mice and ascribed to a misplacement of Cntn5-ablated cells. This phenotype was accompanied by a reduction in the barrel/septa ratio of the S1 barrel field. The structure and morphology of the hippocampus was intact in Cntn5−/− mice. A set of behavioral experiments including social, exploratory and repetitive behaviors showed that these were unaffected in Cntn5−/− mice. Taken together, these data demonstrate a selective role of Cntn5 in development of the cerebral cortex without overt behavioral phenotypes.
Collapse
Affiliation(s)
- Kristel T E Kleijer
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Denise van Nieuwenhuize
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Henk A Spierenburg
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Sara Gregorio-Jordan
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - Martien J H Kas
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| | - J Peter H Burbach
- a Department of Translational Neuroscience , Brain Centre Rudolf Magnus, University Medical Centre Utrecht , Utrecht , the Netherlands
| |
Collapse
|
17
|
Xenopus laevis neuronal cell adhesion molecule (nrcam): plasticity of a CAM in the developing nervous system. Dev Genes Evol 2016; 227:61-67. [PMID: 27942869 DOI: 10.1007/s00427-016-0569-9] [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: 08/03/2016] [Accepted: 11/24/2016] [Indexed: 10/20/2022]
Abstract
Neuron-glial-related cell adhesion molecule (NRCAM) is a neuronal cell adhesion molecule of the L1 immunoglobulin superfamily, which plays diverse roles during nervous system development including axon growth and guidance, synapse formation, and formation of the myelinated nerve. Perturbations in NRCAM function cause a wide variety of disorders, which can affect wiring and targeting of neurons, or cause psychiatric disorders as well as cancers through abnormal modulation of signaling events. In the present study, we characterize the Xenopus laevis homolog of nrcam. Expression of Xenopus nrcam is most abundant along the dorsal midline throughout the developing brain and in the outer nuclear layer of the retina.
Collapse
|
18
|
Zuko A, Oguro-Ando A, van Dijk R, Gregorio-Jordan S, van der Zwaag B, Burbach JPH. Developmental role of the cell adhesion molecule Contactin-6 in the cerebral cortex and hippocampus. Cell Adh Migr 2016; 10:378-92. [PMID: 26939565 DOI: 10.1080/19336918.2016.1155018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The gene encoding the neural cell adhesion molecule Contactin-6 (Cntn6 a.k.a. NB-3) has been implicated as an autism risk gene, suggesting that its mutation is deleterious to brain development. Due to its GPI-anchor at Cntn6 may exert cell adhesion/receptor functions in complex with other membrane proteins, or serve as a ligand. We aimed to uncover novel phenotypes related to Cntn6 functions during development in the cerebral cortex of adult Cntn6(-/-) mice. We first determined Cntn6 protein and mRNA expression in the cortex, thalamic nuclei and the hippocampus at P14, which decreased specifically in the cortex at adult stages. Neuroanatomical analysis demonstrated a significant decrease of Cux1+ projection neurons in layers II-IV and an increase of FoxP2+ projection neurons in layer VI in the visual cortex of adult Cntn6(-/-) mice compared to wild-type controls. Furthermore, the number of parvalbumin+ (PV) interneurons was decreased in Cntn6(-/-) mice, while the amount of NPY+ interneurons remained unchanged. In the hippocampus the delineation and outgrowth of mossy fibers remained largely unchanged, except for the observation of a larger suprapyramidal bundle. The observed abnormalities in the cerebral cortex and hippocampus of Cntn6(-/-) mice suggests that Cntn6 serves developmental functions involving cell survival, migration and fasciculation. Furthermore, these data suggest that Cntn6 engages in both trans- and cis-interactions and may be involved in larger protein interaction networks.
Collapse
Affiliation(s)
- Amila Zuko
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Asami Oguro-Ando
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Roland van Dijk
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Sara Gregorio-Jordan
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Bert van der Zwaag
- b Department of Genetics , University Medical Center Utrecht , Utrecht , The Netherlands
| | - J Peter H Burbach
- a Brain Center Rudolf Magnus , Department of Translational Neuroscience , University Medical Center Utrecht , Utrecht , The Netherlands
| |
Collapse
|
19
|
Kuhn PH, Colombo AV, Schusser B, Dreymueller D, Wetzel S, Schepers U, Herber J, Ludwig A, Kremmer E, Montag D, Müller U, Schweizer M, Saftig P, Bräse S, Lichtenthaler SF. Systematic substrate identification indicates a central role for the metalloprotease ADAM10 in axon targeting and synapse function. eLife 2016; 5. [PMID: 26802628 PMCID: PMC4786429 DOI: 10.7554/elife.12748] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/22/2016] [Indexed: 12/11/2022] Open
Abstract
Metzincin metalloproteases have major roles in intercellular communication by modulating the function of membrane proteins. One of the proteases is the a-disintegrin-and-metalloprotease 10 (ADAM10) which acts as alpha-secretase of the Alzheimer's disease amyloid precursor protein. ADAM10 is also required for neuronal network functions in murine brain, but neuronal ADAM10 substrates are only partly known. With a proteomic analysis of Adam10-deficient neurons we identified 91, mostly novel ADAM10 substrate candidates, making ADAM10 a major protease for membrane proteins in the nervous system. Several novel substrates, including the neuronal cell adhesion protein NrCAM, are involved in brain development. Indeed, we detected mistargeted axons in the olfactory bulb of conditional ADAM10-/- mice, which correlate with reduced cleavage of NrCAM, NCAM and other ADAM10 substrates. In summary, the novel ADAM10 substrates provide a molecular basis for neuronal network dysfunctions in conditional ADAM10-/- mice and demonstrate a fundamental function of ADAM10 in the brain.
Collapse
Affiliation(s)
- Peer-Hendrik Kuhn
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institut für Pathologie und Pathologische Anatomie, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| | - Alessio Vittorio Colombo
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Benjamin Schusser
- Department of Animal Science, Institute for Animal Physiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniela Dreymueller
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Sebastian Wetzel
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Ute Schepers
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Julia Herber
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany
| | - Andreas Ludwig
- Institute of Pharmacology and Toxicology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Elisabeth Kremmer
- German Research Center for Environmental Health, Institute of Molecular Tumor immunology, Helmholtz Zentrum München, Munich, Germany
| | - Dirk Montag
- Neurogenetics, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Ulrike Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michaela Schweizer
- Service-Gruppe für Elektronenmikroskopie, Zentrum für Molekulare Neurobiologie, Hamburg, Germany
| | - Paul Saftig
- Institute of Biochemistry, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Stefan Bräse
- Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Stefan F Lichtenthaler
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
20
|
Kandalepas PC, Vassar R. The normal and pathologic roles of the Alzheimer's β-secretase, BACE1. Curr Alzheimer Res 2015; 11:441-9. [PMID: 24893886 DOI: 10.2174/1567205011666140604122059] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/15/2014] [Accepted: 01/25/2014] [Indexed: 01/18/2023]
Abstract
As the most common neurodegenerative disease, therapeutic avenues for the treatment and prevention of Alzheimer's Disease are highly sought after. The aspartic protease BACE1 is the initiator enzyme for the formation of Aβ, a major constituent of amyloid plaques that represent one of the hallmark pathological features of this disorder. Thus, targeting BACE1 for disease-modifying AD therapies represents a rationale approach. The collective knowledge acquired from investigations of BACE1 deletion mutants and characterization of BACE1 substrates has downstream significance not only for the discovery of AD drug therapies but also for predicting side effects of BACE1 inhibition. Here we discuss the identification and validation of BACE1 as the β-secretase implicated in AD, in addition to information regarding BACE1 cell biology, localization, substrates and potential physiological functions derived from BACE1 knockout models.
Collapse
Affiliation(s)
| | - Robert Vassar
- Northwestern University, Feinberg School of Medicine, Department of Cell & Molecular Biology, 300 E. Superior, Tarry 8-713, IL 60611, Chicago.
| |
Collapse
|
21
|
Abstract
β-site amyloid precursor protein cleaving enzyme 1 (BACE1) is the β-secretase enzyme required for the production of the neurotoxic β-amyloid (Aβ) peptide that is widely considered to have a crucial early role in the etiology of Alzheimer’s disease (AD). As a result, BACE1 has emerged as a prime drug target for reducing the levels of Aβ in the AD brain, and the development of BACE1 inhibitors as therapeutic agents is being vigorously pursued. It has proven difficult for the pharmaceutical industry to design BACE1 inhibitor drugs that pass the blood–brain barrier, however this challenge has recently been met and BACE1 inhibitors are now in human clinical trials to test for safety and efficacy in AD patients and individuals with pre-symptomatic AD. Initial results suggest that some of these BACE1 inhibitor drugs are well tolerated, although others have dropped out because of toxicity and it is still too early to know whether any will be effective for the prevention or treatment of AD. Additionally, based on newly identified BACE1 substrates and phenotypes of mice that lack BACE1, concerns have emerged about potential mechanism-based side effects of BACE1 inhibitor drugs with chronic administration. It is hoped that a therapeutic window can be achieved that balances safety and efficacy. This review summarizes the current state of progress in the development of BACE1 inhibitor drugs and the evaluation of their therapeutic potential for AD.
Collapse
|
22
|
Neural cell adhesion molecule NrCAM is expressed in the mammalian inner ear and modulates spiral ganglion neurite outgrowth in an in vitro alternate choice assay. J Mol Neurosci 2014; 55:836-44. [PMID: 25407819 DOI: 10.1007/s12031-014-0436-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/30/2014] [Indexed: 12/17/2022]
Abstract
Neuron-glial-related cell adhesion molecule (NrCAM) is a neuronal cell adhesion molecule involved in neuron-neuron and neuron-glial adhesion as well as directional signaling during axonal cone growth. NrCAM has been shown to be involved in several cellular processes in the central and peripheral nervous systems, including neurite outgrowth, axonal pathfinding and myelination, fasciculation of nerve fibers, and cell migration. This includes sensory systems such as the eye and olfactory system. However, there are no reports on the expression/function of NrCAM in the auditory system. The aim of the present study was to elucidate the occurrence of NrCAM in the mammalian cochlea and its role in innervation of the auditory end organ. Our work indicates that NrCAM is highly expressed in the developing mammalian cochlea (position consistent with innervation). Moreover, we found that NrCAM, presented in stripe micropatterns, provide directional cues to neonatal rat inner ear spiral ganglion neurites in vitro. Our results are consistent with a role for NrCAM in the pathfinding of spiral ganglion dendrites toward their hair cell targets in the sensory epithelium.
Collapse
|
23
|
Van Battum EY, Gunput RAF, Lemstra S, Groen EJN, Yu KL, Adolfs Y, Zhou Y, Hoogenraad CC, Yoshida Y, Schachner M, Akhmanova A, Pasterkamp RJ. The intracellular redox protein MICAL-1 regulates the development of hippocampal mossy fibre connections. Nat Commun 2014; 5:4317. [PMID: 25007825 DOI: 10.1038/ncomms5317] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/05/2014] [Indexed: 02/05/2023] Open
Abstract
Mical is a reduction-oxidation (redox) enzyme that functions as an unusual F-actin disassembly factor during Drosophila development. Although three Molecule interacting with CasL (MICAL) proteins exist in vertebrate species, their mechanism of action remains poorly defined and their role in vivo unknown. Here, we report that vertebrate MICAL-1 regulates the targeting of secretory vesicles containing immunoglobulin superfamily cell adhesion molecules (IgCAMs) to the neuronal growth cone membrane through its ability to control the actin cytoskeleton using redox chemistry, thereby maintaining appropriate IgCAM cell surface levels. This precise regulation of IgCAMs by MICAL-1 is essential for the lamina-specific targeting of mossy fibre axons onto CA3 pyramidal neurons in the developing mouse hippocampus in vivo. These findings reveal the first in vivo role for a vertebrate MICAL protein, expand the repertoire of cellular functions controlled through MICAL-mediated effects on the cytoskeleton, and provide insights into the poorly characterized mechanisms underlying neuronal protein cell surface expression and lamina-specific axonal targeting.
Collapse
Affiliation(s)
- Eljo Y Van Battum
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2]
| | - Rou-Afza F Gunput
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2] [3]
| | - Suzanne Lemstra
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Ewout J N Groen
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2] Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3508 GA, Utrecht, The Netherlands
| | - Ka Lou Yu
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| | - Yeping Zhou
- 1] Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands [2]
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Yukata Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
24
|
Vassar R, Kuhn PH, Haass C, Kennedy ME, Rajendran L, Wong PC, Lichtenthaler SF. Function, therapeutic potential and cell biology of BACE proteases: current status and future prospects. J Neurochem 2014; 130:4-28. [PMID: 24646365 PMCID: PMC4086641 DOI: 10.1111/jnc.12715] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/12/2014] [Accepted: 03/14/2014] [Indexed: 01/18/2023]
Abstract
The β-site APP cleaving enzymes 1 and 2 (BACE1 and BACE2) were initially identified as transmembrane aspartyl proteases cleaving the amyloid precursor protein (APP). BACE1 is a major drug target for Alzheimer's disease because BACE1-mediated cleavage of APP is the first step in the generation of the pathogenic amyloid-β peptides. BACE1, which is highly expressed in the nervous system, is also required for myelination by cleaving neuregulin 1. Several recent proteomic and in vivo studies using BACE1- and BACE2-deficient mice demonstrate a much wider range of physiological substrates and functions for both proteases within and outside of the nervous system. For BACE1 this includes axon guidance, neurogenesis, muscle spindle formation, and neuronal network functions, whereas BACE2 was shown to be involved in pigmentation and pancreatic β-cell function. This review highlights the recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer's disease. The protease BACE1 is a major drug target in Alzheimer disease. Together with its homolog BACE2, both proteases have an increasing number of functions within and outside of the nervous system. This review highlights recent progress in understanding cell biology, substrates, and functions of BACE proteases and discusses the therapeutic options and potential mechanism-based liabilities, in particular for BACE inhibitors in Alzheimer disease.
Collapse
Affiliation(s)
- Robert Vassar
- Department of Cell and Molecular Biology, Feinberg University School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peer-Hendrik Kuhn
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Adolf-Butenandt Institute, Biochemistry, Ludwig-Maximilians University Munich, Munich, Germany
| | - Matthew E. Kennedy
- Neurosciences, Merck Research Labs, Boston, Massachusetts, USA
- Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, Division of Psychiatry Research, University of Zurich, Zurich, Switzerland
- Graduate programs of the Zurich Center for Integrative Human Physiology and Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| | - Philip C. Wong
- Departments of Pathology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
25
|
Abstract
The β secretase, widely known as β-site amyloid precursor protein cleaving enzyme 1 (BACE1), initiates the production of the toxic amyloid β (Aβ) that plays a crucial early part in Alzheimer's disease pathogenesis. BACE1 is a prime therapeutic target for lowering cerebral Aβ concentrations in Alzheimer's disease, and clinical development of BACE1 inhibitors is being intensely pursued. Although BACE1 inhibitor drug development has proven challenging, several promising BACE1 inhibitors have recently entered human clinical trials. The safety and efficacy of these drugs are being tested at present in healthy individuals and patients with Alzheimer's disease, and will soon be tested in individuals with presymptomatic Alzheimer's disease. Although hopes are high that BACE1 inhibitors might be efficacious for the prevention or treatment of Alzheimer's disease, concerns have been raised about potential mechanism-based side-effects of these drugs. The potential of therapeutic BACE1 inhibition might prove to be a watershed in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Riqiang Yan
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robert Vassar
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
26
|
Qiu XS, Lv F, Zhu ZZ, Qian BP, Wang B, Yu Y, Qiu Y. Lack of association between the CHL1 gene and adolescent idiopathic scoliosis susceptibility in Han Chinese: a case-control study. BMC Musculoskelet Disord 2014; 15:38. [PMID: 24512353 PMCID: PMC3925962 DOI: 10.1186/1471-2474-15-38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 02/06/2014] [Indexed: 11/20/2022] Open
Abstract
Background A previous genome-wide association study (GWAS) suggested a strong association between the single nucleotide polymorphism (SNP) rs10510181 in the proximity of the gene encoding a cell adhesion molecule with homology to L1CAM (CHL1) and adolescent idiopathic scoliosis (AIS) in Caucasians. To clarify the role of CHL1 in the etiopathogenesis of AIS, we performed a case-control replication study in a Han Chinese population. Methods Five hundred female AIS patients between 10 and 18 years of age, as well as 500 age- and sex-matched controls were included. This study was conducted as a 2-stage case-control analysis: initial screening for the association between AIS and SNPs in and around the CHL1 gene (186 cases and 169 controls) followed by a confirmation test (314 cases and 331 controls). rs10510181 and 4 SNPs (rs2055314, rs331894, rs2272522, and rs2272524) in the CHL1 gene were selected for genotyping. Results Putative associations were shown between AIS and rs10510181, rs2055314, and rs2272522 in stage I. However, the associations were not confirmed in stage II. For rs10510181, the genotype frequencies were GG 28.8%, GA 46.2%, and AA 25.0% in AIS patients and GG 29.8%, GA 48.8%, and AA 21.4% in controls. No significant difference was found in genotype distribution between cases and controls (P = 0.39). Similarly, the genotype and allele distribution were comparable between case and control for rs2055314 and rs2272522. Conclusions There was no statistical association between polymorphisms of the CHL1 gene and idiopathic scoliosis in a Chinese population.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yong Qiu
- Spine Surgery, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
27
|
Nagaraj K, Mualla R, Hortsch M. The L1 Family of Cell Adhesion Molecules: A Sickening Number of Mutations and Protein Functions. ADVANCES IN NEUROBIOLOGY 2014; 8:195-229. [DOI: 10.1007/978-1-4614-8090-7_9] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
28
|
The Alzheimer's β-secretase BACE1 localizes to normal presynaptic terminals and to dystrophic presynaptic terminals surrounding amyloid plaques. Acta Neuropathol 2013; 126:329-52. [PMID: 23820808 PMCID: PMC3753469 DOI: 10.1007/s00401-013-1152-3] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 06/20/2013] [Indexed: 01/18/2023]
Abstract
β-Site amyloid precursor protein (APP) cleaving enzyme-1 (BACE1) is the β-secretase that initiates Aβ production in Alzheimer’s disease (AD). BACE1 levels are increased in AD, which could contribute to pathogenesis, yet the mechanism of BACE1 elevation is unclear. Furthermore, the normal function of BACE1 is poorly understood. We localized BACE1 in the brain at both the light and electron microscopic levels to gain insight into normal and pathophysiologic roles of BACE1 in health and AD, respectively. Our findings provide the first ultrastructural evidence that BACE1 localizes to vesicles (likely endosomes) in normal hippocampal mossy fiber terminals of both non-transgenic and APP transgenic (5XFAD) mouse brains. In some instances, BACE1-positive vesicles were located near active zones, implying a function for BACE1 at the synapse. In addition, BACE1 accumulated in swollen dystrophic autophagosome-poor presynaptic terminals surrounding amyloid plaques in 5XFAD cortex and hippocampus. Importantly, accumulations of BACE1 and APP co-localized in presynaptic dystrophies, implying increased BACE1 processing of APP in peri-plaque regions. In primary cortical neuron cultures, treatment with the lysosomal protease inhibitor leupeptin caused BACE1 levels to increase; however, exposure of neurons to the autophagy inducer trehalose did not reduce BACE1 levels. This suggests that BACE1 is degraded by lysosomes but not by autophagy. Our results imply that BACE1 elevation in AD could be linked to decreased lysosomal degradation of BACE1 within dystrophic presynaptic terminals. Elevated BACE1 and APP levels in plaque-associated presynaptic dystrophies could increase local peri-plaque Aβ generation and accelerate amyloid plaque growth in AD.
Collapse
|
29
|
Hitt B, Riordan SM, Kukreja L, Eimer WA, Rajapaksha TW, Vassar R. β-Site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1)-deficient mice exhibit a close homolog of L1 (CHL1) loss-of-function phenotype involving axon guidance defects. J Biol Chem 2012; 287:38408-25. [PMID: 22988240 DOI: 10.1074/jbc.m112.415505] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACE1 is the β-secretase enzyme that initiates production of the β-amyloid peptide involved in Alzheimer disease. However, little is known about the functions of BACE1. BACE1-deficient mice exhibit mild but complex neurological phenotypes suggesting therapeutic BACE1 inhibition may not be completely free of mechanism-based side effects. Recently, we have reported that BACE1 null mice have axon guidance defects in olfactory sensory neuron projections to glomeruli in the olfactory bulb. Here, we show that BACE1 deficiency also causes an axon guidance defect in the hippocampus, a shortened and disorganized infrapyramidal bundle of the mossy fiber projection from the dentate gyrus to CA3. Although we observed that a classical axon guidance molecule, EphA4, was cleaved by BACE1 when co-expressed with BACE1 in HEK293 cells, we could find no evidence of BACE1 processing of EphA4 in the brain. Remarkably, we discovered that the axon guidance defects of BACE1(-/-) mice were strikingly similar to those of mice deficient in a recently identified BACE1 substrate, the neural cell adhesion molecule close homolog of L1 (CHL1) that is involved in neurite outgrowth. CHL1 undergoes BACE1-dependent processing in BACE1(+/+), but not BACE1(-/-), hippocampus, and olfactory bulb, indicating that CHL1 is a BACE1 substrate in vivo. Finally, BACE1 and CHL1 co-localize in the terminals of hippocampal mossy fibers, olfactory sensory neuron axons, and growth cones of primary hippocampal neurons. We conclude that BACE1(-/-) axon guidance defects are likely the result of abrogated BACE1 processing of CHL1 and that BACE1 deficiency produces a CHL1 loss-of-function phenotype. Our results imply the possibility that axon mis-targeting may occur in adult neurogenic and/or regenerating neurons as a result of chronic BACE1 inhibition and add a note of caution to BACE1 inhibitor development.
Collapse
Affiliation(s)
- Brian Hitt
- Department of Cell and Molecular Biology, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
30
|
Sakurai T. The role of NrCAM in neural development and disorders--beyond a simple glue in the brain. Mol Cell Neurosci 2011; 49:351-63. [PMID: 22182708 DOI: 10.1016/j.mcn.2011.12.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 11/03/2011] [Accepted: 12/02/2011] [Indexed: 12/15/2022] Open
Abstract
NrCAM is a neuronal cell adhesion molecule of the L1 family of immunoglobulin super family. It plays a wide variety of roles in neural development, including cell proliferation and differentiation, axon growth and guidance, synapse formation, and the formation of the myelinated nerve structure. NrCAM functions in cell adhesion and modulates signaling pathways in neural development through multiple molecular interactions with guidance and other factors. Alterations in NrCAM structure/expression are associated with psychiatric disorders such as autism and drug addiction and with tumor progression. The mechanisms of NrCAM participation in development and how these might be perturbed in disorders are reviewed.
Collapse
Affiliation(s)
- Takeshi Sakurai
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Japan.
| |
Collapse
|
31
|
van Velthoven CTJ, Kavelaars A, van Bel F, Heijnen CJ. Mesenchymal stem cell transplantation changes the gene expression profile of the neonatal ischemic brain. Brain Behav Immun 2011; 25:1342-8. [PMID: 21473911 DOI: 10.1016/j.bbi.2011.03.021] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 03/29/2011] [Accepted: 03/31/2011] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cell (MSC) treatment is an effective strategy to reduce brain damage after neonatal hypoxia-ischemia (HI) in mice. We recently showed that a single injection with MSC at either 3 or 10 days after HI (MSC-3 or MSC-10) increases neurogenesis. In case of two injections (MSC-3+10), the second MSC application does not increase neurogenesis, but promotes corticospinal tract remodeling. Here we investigated GFP(+)-MSC engraftment level in the brain using quantitative-PCR analysis. We show for the first time that in the neonatal ischemic brain survival of transplanted MSC is very limited. At 3 days after injection ∼22% of transplanted MSC were still detectable and 18 days after the last administration barely ∼1%. These findings indicate that engraftment of MSC is not likely the underlying mechanism of the efficient regenerative process. Therefore, we tested the hypothesis that the effects of MSC-treatment on regenerative processes are related to specific changes in the gene expression of growth factors and cytokines in the damaged area of the brain using PCR-array analysis. We compared the effect of one (MSC-10) or two (MSC-3+10) injections of 10(5) MSC on gene expression in the brain. Our data show that MSC-10 induced expression of genes regulating proliferation/survival. In response to MSC-3+10-treatment a pattern functionally categorized as growth stimulating genes was increased. Collectively, our data indicate that specific regulation of the endogenous growth factor milieu rather than replacement of damaged tissue by exogenous MSC mediates regeneration of the damaged neonatal brain by MSC-treatment.
Collapse
Affiliation(s)
- Cindy T J van Velthoven
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | | | | | | |
Collapse
|
32
|
Messenger RNA and microRNA profiling during early mouse EB formation. Gene Expr Patterns 2011; 11:334-44. [PMID: 21440681 DOI: 10.1016/j.gep.2011.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 02/21/2011] [Accepted: 03/14/2011] [Indexed: 11/22/2022]
Abstract
Embryonic stem (ES) cells can be induced to differentiate into embryoid bodies (EBs) in a synchronised manner when plated at a fixed density in hanging drops. This differentiation procedure mimics post-implantation development in mouse embryos and also serves as the starting point of protocols used in differentiation of stem cells into various lineages. Currently, little is known about the potential influence of microRNAs (miRNAs) on mRNA expression patterns during EB formation. We have measured mRNA and miRNA expression in developing EBs plated in hanging drops until day 3, when discrete structural changes occur involving their differentiation into three germ layers. We observe significant alterations in mRNA and miRNA expression profiles during this early developmental time frame, in particular of genes involved in germ layer formation, stem cell pluripotency and nervous system development. Computational target prediction using Pictar, TargetScan and miRBase Targets reveals an enrichment of binding sites corresponding to differentially and highly expressed miRNAs in stem cell pluripotency genes and a neuroectodermal marker, Nes. We also find that members of let-7 family are significantly down-regulated at day 3 and the corresponding up-regulated genes are enriched in let-7 seed sequences. These results depict how miRNA expression changes may affect the expression of mRNAs involved in EB formation on a genome-wide scale. Understanding the regulatory effects of miRNAs during EB formation may enable more efficient derivation of different cell types in culture.
Collapse
|
33
|
Xenaki D, Martin IB, Yoshida L, Ohyama K, Gennarini G, Grumet M, Sakurai T, Furley AJW. F3/contactin and TAG1 play antagonistic roles in the regulation of sonic hedgehog-induced cerebellar granule neuron progenitor proliferation. Development 2011; 138:519-29. [PMID: 21205796 DOI: 10.1242/dev.051912] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Modulation of the sonic hedgehog (SHH) pathway is a crucial factor in cerebellar morphogenesis. Stimulation of granule neuron progenitor (GNP) proliferation is a central function of SHH signalling, but how this is controlled locally is not understood. We show that two sequentially expressed members of the contactin (CNTN) family of adhesion molecules, TAG1 and F3, act antagonistically to control SHH-induced proliferation: F3 suppresses SHH-induced GNP proliferation and induces differentiation, whereas TAG1 antagonises F3. Production of GNPs in TAG1-null mice is delayed and reduced. F3 and TAG1 colocalise on GNPs with the related L1-like adhesion molecule NrCAM, and F3 fails to suppress the SHH-induced proliferation of NrCAM-deficient GNPs. We show that F3 and SHH both primarily affect a group of intermediate GNPs (IPs), which, though actively dividing, also express molecules associated with differentiation, including β-tubulin III (TuJ1) and TAG1. In vivo, intermediate progenitors form a discrete layer in the middle of the external germinal layer (mEGL), while F3 becomes expressed on the axons of postmitotic granule neurons as they leave the inner EGL (iEGL). We propose, therefore, that F3 acts as a localised signal in the iEGL that induces SHH-stimulated cells in the overlying mEGL to exit cell cycle and differentiate. By contrast, expression of TAG1 on GNPs antagonises this signal in the mEGL, preventing premature differentiation and sustaining GNP expansion in a paracrine fashion. Together, these findings indicate that CNTN and L1-like proteins play a significant role in modulating SHH-induced neuronal precursor proliferation.
Collapse
Affiliation(s)
- Dia Xenaki
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The dystrophin protein complex, an important regulator of muscle membrane integrity, also maintains neural organization through interactions with the L1CAM family member SAX-7. The dystrophin protein complex (DPC), composed of dystrophin and associated proteins, is essential for maintaining muscle membrane integrity. The link between mutations in dystrophin and the devastating muscle failure of Duchenne’s muscular dystrophy (DMD) has been well established. Less well appreciated are the accompanying cognitive impairment and neuropsychiatric disorders also presented in many DMD patients, which suggest a wider role for dystrophin in membrane–cytoskeleton function. This study provides genetic evidence of a novel role for DYS-1/dystrophin in maintaining neural organization in Caenorhabditis elegans. This neuronal function is distinct from the established role of DYS-1/dystrophin in maintaining muscle integrity and regulating locomotion. SAX-7, an L1 cell adhesion molecule (CAM) homologue, and STN-2/γ-syntrophin also function to maintain neural integrity in C. elegans. This study provides biochemical data that show that SAX-7 associates with DYS-1 in an STN-2/γ-syntrophin–dependent manner. These results reveal a recruitment of L1CAMs to the DPC to ensure neural integrity is maintained.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Genetics, Cell Biology, and Development, Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
35
|
Moy SS, Nonneman RJ, Young NB, Demyanenko GP, Maness PF. Impaired sociability and cognitive function in Nrcam-null mice. Behav Brain Res 2009; 205:123-31. [PMID: 19540269 PMCID: PMC2753746 DOI: 10.1016/j.bbr.2009.06.021] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2009] [Revised: 05/12/2009] [Accepted: 06/10/2009] [Indexed: 11/24/2022]
Abstract
NRCAM (Neuronal Cell Adhesion Molecule) has an important role in axonal guidance and the organization of neural circuitry during brain development. Association analyses in human populations have identified NRCAM as a candidate gene for autism susceptibility. In the present study, we evaluated Nrcam-null mice for sociability, social novelty preference, and reversal learning as a model for the social deficits, repetitive behavior, and cognitive rigidity characteristic of autism. Prepulse inhibition of acoustic startle responses was also measured, to reflect sensorimotor-gating deficits in autism spectrum disorders. Assays for anxiety-like behavior in an elevated plus maze and open field, motor coordination, and olfactory ability in a buried food test were conducted to provide control measures for the interpretation of results. Overall, the loss of Nrcam led to behavioral alterations in sociability, acquisition of a spatial task, and reversal learning, dependent on sex. In comparison to male wild type mice, male Nrcam-null mutants had significantly decreased sociability in a three-chambered choice task. Low sociability in the male null mutants was not associated with changes in anxiety-like behavior, activity, or motor coordination. Male, but not female, Nrcam-null mice had small decreases in prepulse inhibition. Nrcam deficiency in female mice led to impaired acquisition of spatial learning in the Morris water maze task. Reversal learning deficits were observed in both male and female Nrcam-null mice. These results provide evidence that NRCAM mediates domains of function relevant to symptoms observed in autism.
Collapse
Affiliation(s)
- Sheryl S Moy
- Neurodevelopmental Disorders Research Center, CB#7146, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA.
| | | | | | | | | |
Collapse
|
36
|
de Castro F. Wiring Olfaction: The Cellular and Molecular Mechanisms that Guide the Development of Synaptic Connections from the Nose to the Cortex. Front Neurosci 2009; 3:52. [PMID: 20582279 PMCID: PMC2858608 DOI: 10.3389/neuro.22.004.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 11/04/2009] [Indexed: 12/27/2022] Open
Abstract
Within the central nervous system, the olfactory system fascinates by its developmental and physiological particularities, and is one of the most studied models to understand the mechanisms underlying the guidance of growing axons to their appropriate targets. A constellation of contact-mediated (laminins, CAMs, ephrins, etc.) and secreted mechanisms (semaphorins, slits, growth factors, etc.) are known to play different roles in the establishment of synaptic interactions between the olfactory epithelium, olfactory bulb (OB) and olfactory cortex. Specific mechanisms of this system (including the amazing family of about 1000 different olfactory receptors) have been also proposed. In the last years, different reviews have focused in partial sights, specially in the mechanisms involved in the formation of the olfactory nerve, but a detailed review of the mechanisms implicated in the development of the connections among the different olfactory structures (olfactory epithelium, OB, olfactory cortex) remains to be written. In the present work, we afford this systematic review: the different cellular and molecular mechanisms which rule the formation of the olfactory nerve, the lateral olfactory tract and the intracortical connections, as well as the few data available regarding the accessory olfactory system. These mechanisms are compared, and the implications of the differences and similarities discussed in this fundamental scenario of ontogeny.
Collapse
Affiliation(s)
- Fernando de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos Toledo, Spain
| |
Collapse
|
37
|
Nagaraj K, Kristiansen LV, Skrzynski A, Castiella C, Garcia-Alonso L, Hortsch M. Pathogenic human L1-CAM mutations reduce the adhesion-dependent activation of EGFR. Hum Mol Genet 2009; 18:3822-31. [PMID: 19617634 PMCID: PMC2748892 DOI: 10.1093/hmg/ddp325] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/08/2009] [Accepted: 07/16/2009] [Indexed: 11/13/2022] Open
Abstract
L1-cell adhesion molecule (L1-CAM) belongs to a functionally conserved group of neural cell adhesion molecules that are implicated in many aspects of nervous system development. In many neuronal cells the adhesive function of L1-type CAMs induces cellular signaling processes that involves the activation of neuronal tyrosine protein kinases and among other functions regulates axonal growth and guidance. Mutations in the human L1-CAM gene are responsible for a complex neurodevelopmental condition, generally referred to as L1 syndrome. Several pathogenic L1-CAM mutations have been identified in humans that cause L1 syndrome in affected individuals without affecting the level of L1-CAM-mediated homophilic cell adhesion when tested in vitro. In this study, an analysis of two different pathogenic human L1-CAM molecules indicates that although both induce normal L1-CAM-mediated cell aggregation, they are defective in stimulating human epidermal growth factor receptor tyrosine kinase activity in vitro and are unable to rescue L1 loss-of-function conditions in a Drosophila transgenic model in vivo. These results indicate that the L1 syndrome-associated phenotype might involve the disruption of L1-CAM's functions at different levels. Either by reducing or abolishing L1-CAM protein expression, by interfering with L1-CAM's cell surface expression, by reducing L1-CAM's adhesive ability or by impeding further downstream adhesion-dependent signaling processes.
Collapse
Affiliation(s)
- Kakanahalli Nagaraj
- Department of Cell and Developmental Biology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Lars V. Kristiansen
- Department of Cell and Developmental Biology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
- The Protein Laboratory, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark and
- Instituto de Neurociencias CSIC-UMH, Universidad Miguel Hernandez, 03550 Sant Joan d'Alacant, Spain
| | - Adam Skrzynski
- Department of Cell and Developmental Biology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | - Carlos Castiella
- Instituto de Neurociencias CSIC-UMH, Universidad Miguel Hernandez, 03550 Sant Joan d'Alacant, Spain
| | - Luis Garcia-Alonso
- Instituto de Neurociencias CSIC-UMH, Universidad Miguel Hernandez, 03550 Sant Joan d'Alacant, Spain
| | - Michael Hortsch
- Department of Cell and Developmental Biology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
38
|
Katidou M, Vidaki M, Strigini M, Karagogeos D. The immunoglobulin superfamily of neuronal cell adhesion molecules: lessons from animal models and correlation with human disease. Biotechnol J 2009; 3:1564-80. [PMID: 19072911 DOI: 10.1002/biot.200800281] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neuronal cell adhesion molecules of the immunoglobulin superfamily (IgCAMs) play a crucial role in the formation of neural circuits at different levels: cell migration, axonal and dendritic targeting as well as synapse formation. Furthermore, in perinatal and adult life, neuronal IgCAMs are required for the formation and maintenance of specialized axonal membrane domains, synaptic plasticity and neurogenesis. Mutations in the corresponding human genes have been correlated to several human neuronal disorders. Perturbing neuronal IgCAMs in animal models provides powerful means to understand the molecular and cellular basis of such human disorders. In this review, we concentrate on the NCAM, L1 and contactin subfamilies of neuronal IgCAMs summarizing recent functional studies from model systems and highlighting their links to disease pathogenesis.
Collapse
Affiliation(s)
- Markella Katidou
- University of Crete, Faculty of Medicine, Vassilika Vouton, Heraklion, Crete, Greece
| | | | | | | |
Collapse
|