1
|
Barman B, Thakur MK. Neuropsin promotes hippocampal synaptogenesis by regulating the expression and cleavage of L1CAM. J Cell Sci 2024; 137:jcs261422. [PMID: 38206094 DOI: 10.1242/jcs.261422] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
During early postnatal brain development, the formation of proper synaptic connections between neurons is crucial for the development of functional neural networks. Recent studies have established the involvement of protease-mediated modulations of extracellular components in both synapse formation and elimination. The secretory serine protease neuropsin (also known as kallikrein-8) cleaves a few transmembrane or extracellular matrix proteins in a neural activity-dependent manner and regulates neural plasticity. However, neuropsin-dependent proteolysis of extracellular components and the involvement of these components in mouse brain development are poorly understood. We have observed that during hippocampus development, expression of neuropsin and levels of full-length or cleaved fragments of the neuropsin substrate protein L1 cell adhesion molecule (L1CAM) positively correlate with synaptogenesis. Our subcellular fractionation studies show that the expression of neuropsin and its proteolytic activity on L1CAM are enriched at developing hippocampal synapses. Activation of neuropsin expression upregulates the transcription and cleavage of L1CAM. Furthermore, blocking of neuropsin activity, as well as knockdown of L1CAM expression, significantly downregulates in vitro hippocampal synaptogenesis. Taken together, these findings provide evidence for the involvement of neuropsin activity-dependent regulation of L1CAM expression and cleavage in hippocampal synaptogenesis.
Collapse
Affiliation(s)
- Bhabotosh Barman
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Mahendra Kumar Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
2
|
Duan W, Xia S, Tang M, Lin M, Liu W, Wang Q. Targeting of endothelial cells in brain tumours. Clin Transl Med 2023; 13:e1433. [PMID: 37830128 PMCID: PMC10570772 DOI: 10.1002/ctm2.1433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Aggressive brain tumours, whether primary gliomas or secondary metastases, are characterised by hypervascularisation and are fatal. Recent research has emphasised the crucial involvement of endothelial cells (ECs) in all brain tumour genesis and development events, with various patterns and underlying mechanisms identified. MAIN BODY Here, we highlight recent advances in knowledge about the contributions of ECs to brain tumour development, providing a comprehensive summary including descriptions of interactions between ECs and tumour cells, the heterogeneity of ECs and new models for research on ECs in brain malignancies. We also discuss prospects for EC targeting in novel therapeutic approaches. CONCLUSION Interventions targeting ECs, as an adjunct to other therapies (e.g. immunotherapies, molecular-targeted therapies), have shown promising clinical efficacy due to the high degree of vascularisation in brain tumours. Developing precise strategies to target tumour-associated vessels based on the heterogeneity of ECs is expected to improve anti-vascular efficacy.
Collapse
Affiliation(s)
- Wenzhe Duan
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Shengkai Xia
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Mengyi Tang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Manqing Lin
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
| | - Wenwen Liu
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| | - Qi Wang
- Department of Respiratory MedicineThe Second HospitalDalian Medical UniversityDalianChina
- Cancer Translational Medicine Research CenterThe Second HospitalDalian Medical UniversityDalianChina
| |
Collapse
|
3
|
Congiu L, Granato V, Jakovcevski I, Kleene R, Fernandes L, Freitag S, Kneussel M, Schachner M, Loers G. Mice Mutated in the Third Fibronectin Domain of L1 Show Enhanced Hippocampal Neuronal Cell Death, Astrogliosis and Alterations in Behavior. Biomolecules 2023; 13:776. [PMID: 37238646 PMCID: PMC10216033 DOI: 10.3390/biom13050776] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Adhesion molecules play major roles in cell proliferation, migration, survival, neurite outgrowth and synapse formation during nervous system development and in adulthood. The neural cell adhesion molecule L1 contributes to these functions during development and in synapse formation and synaptic plasticity after trauma in adulthood. Mutations of L1 in humans result in L1 syndrome, which is associated with mild-to-severe brain malformations and mental disabilities. Furthermore, mutations in the extracellular domain were shown to cause a severe phenotype more often than mutations in the intracellular domain. To explore the outcome of a mutation in the extracellular domain, we generated mice with disruption of the dibasic sequences RK and KR that localize to position 858RKHSKR863 in the third fibronectin type III domain of murine L1. These mice exhibit alterations in exploratory behavior and enhanced marble burying activity. Mutant mice display higher numbers of caspase 3-positive neurons, a reduced number of principle neurons in the hippocampus, and an enhanced number of glial cells. Experiments suggest that disruption of the dibasic sequence in L1 results in subtle impairments in brain structure and functions leading to obsessive-like behavior in males and reduced anxiety in females.
Collapse
Affiliation(s)
- Ludovica Congiu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Viviana Granato
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Igor Jakovcevski
- Institut für Anatomie und Klinische Morphologie, Universität Witten/Herdecke, 58455 Witten, Germany;
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Luciana Fernandes
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Sandra Freitag
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Matthias Kneussel
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Falkenried 94, 20251 Hamburg, Germany (R.K.); (S.F.); (M.K.)
| |
Collapse
|
4
|
Hodges SL, Bouza AA, Isom LL. Therapeutic Potential of Targeting Regulated Intramembrane Proteolysis Mechanisms of Voltage-Gated Ion Channel Subunits and Cell Adhesion Molecules. Pharmacol Rev 2022; 74:1028-1048. [PMID: 36113879 PMCID: PMC9553118 DOI: 10.1124/pharmrev.121.000340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/13/2022] [Indexed: 10/03/2023] Open
Abstract
Several integral membrane proteins undergo regulated intramembrane proteolysis (RIP), a tightly controlled process through which cells transmit information across and between intracellular compartments. RIP generates biologically active peptides by a series of proteolytic cleavage events carried out by two primary groups of enzymes: sheddases and intramembrane-cleaving proteases (iCLiPs). Following RIP, fragments of both pore-forming and non-pore-forming ion channel subunits, as well as immunoglobulin super family (IgSF) members, have been shown to translocate to the nucleus to function in transcriptional regulation. As an example, the voltage-gated sodium channel β1 subunit, which is also an IgSF-cell adhesion molecule (CAM), is a substrate for RIP. β1 RIP results in generation of a soluble intracellular domain, which can regulate gene expression in the nucleus. In this review, we discuss the proposed RIP mechanisms of voltage-gated sodium, potassium, and calcium channel subunits as well as the roles of their generated proteolytic products in the nucleus. We also discuss other RIP substrates that are cleaved by similar sheddases and iCLiPs, such as IgSF macromolecules, including CAMs, whose proteolytically generated fragments function in the nucleus. Importantly, dysfunctional RIP mechanisms are linked to human disease. Thus, we will also review how understanding RIP events and subsequent signaling processes involving ion channel subunits and IgSF proteins may lead to the discovery of novel therapeutic targets. SIGNIFICANCE STATEMENT: Several ion channel subunits and immunoglobulin superfamily molecules have been identified as substrates of regulated intramembrane proteolysis (RIP). This signal transduction mechanism, which generates polypeptide fragments that translocate to the nucleus, is an important regulator of gene transcription. RIP may impact diseases of excitability, including epilepsy, cardiac arrhythmia, and sudden death syndromes. A thorough understanding of the role of RIP in gene regulation is critical as it may reveal novel therapeutic strategies for the treatment of previously intractable diseases.
Collapse
Affiliation(s)
- Samantha L Hodges
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Alexandra A Bouza
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| | - Lori L Isom
- Departments of Pharmacology (S.L.H., A.A.B., L.L.I.), Neurology (L.L.I.), and Molecular & Integrative Physiology (L.L.I.), University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
5
|
Stoyanova II, Lutz D. Functional Diversity of Neuronal Cell Adhesion and Recognition Molecule L1CAM through Proteolytic Cleavage. Cells 2022; 11:cells11193085. [PMID: 36231047 PMCID: PMC9562852 DOI: 10.3390/cells11193085] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
The neuronal cell adhesion and recognition molecule L1 does not only 'keep cells together' by way of homophilic and heterophilic interactions, but can also promote cell motility when cleaved into fragments by several proteases. It has largely been thought that such fragments are signs of degradation. Now, it is clear that proteolysis contributes to the pronounced functional diversity of L1, which we have reviewed in this work. L1 fragments generated at the plasma membrane are released into the extracellular space, whereas other membrane-bound fragments are internalised and enter the nucleus, thus conveying extracellular signals to the cell interior. Post-translational modifications on L1 determine the sequence of cleavage by proteases and the subcellular localisation of the generated fragments. Inside the neuronal cells, L1 fragments interact with various binding partners to facilitate morphogenic events, as well as regenerative processes. The stimulation of L1 proteolysis via injection of L1 peptides or proteases active on L1 or L1 mimetics is a promising tool for therapy of injured nervous systems. The collective findings gathered over the years not only shed light on the great functional diversity of L1 and its fragments, but also provide novel mechanistic insights into the adhesion molecule proteolysis that is active in the developing and diseased nervous system.
Collapse
Affiliation(s)
- Irina I. Stoyanova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University, 9002 Varna, Bulgaria
- Department of Brain Ischemia Mechanisms, Research Institute, Medical University, 9002 Varna, Bulgaria
- Correspondence: (I.I.S.); (D.L.)
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum,
44801 Bochum, Germany
- Correspondence: (I.I.S.); (D.L.)
| |
Collapse
|
6
|
Kleene R, Loers G, Castillo G, Schachner M. Cell adhesion molecule L1 interacts with the chromo shadow domain of heterochromatin protein 1 isoforms α, β, and ɣ via its intracellular domain. FASEB J 2021; 36:e22074. [PMID: 34859928 DOI: 10.1096/fj.202100816r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/01/2021] [Accepted: 11/15/2021] [Indexed: 11/11/2022]
Abstract
Cell adhesion molecule L1 regulates multiple cell functions and L1 deficiency is linked to several neural diseases. Proteolytic processing generates functionally decisive L1 fragments, which are imported into the nucleus. By computational analysis, we found at L1's C-terminal end the chromo shadow domain-binding motif PxVxL, which directs the binding of nuclear proteins to the heterochromatin protein 1 (HP1) isoforms α, β, and ɣ. By enzyme-linked immunosorbent assay, we show that the intracellular L1 domain binds to all HP1 isoforms. These interactions involve the HP1 chromo shadow domain and are mediated via the sequence 1158 KDET1161 in the intracellular domain of murine L1, but not by L1's C-terminal PxVxL motif. Immunoprecipitation using nuclear extracts from the brain and from cultured cerebellar and cortical neurons indicates that HP1 isoforms interact with a yet unknown nuclear L1 fragment of approximately 55 kDa (L1-55), which carries ubiquitin residues. Proximity ligation indicates a close association between L1-55 and the HP1 isoforms in neuronal nuclei. This association is reduced after the treatment of neurons with inhibitors of metalloproteases, β-site of amyloid precursor protein cleaving enzyme (BACE1), or ɣ-secretase, suggesting that cleavage of full-length L1 by these proteases generates L1-55. Reduction of HP1α, -β, or -ɣ expression by siRNA decreases L1-dependent neurite outgrowth from cultured cortical neurons and decreases the L1-dependent migration of L1-transfected HEK293 cells in a scratch assay. These findings indicate that the interaction of the novel fragment L1-55 with HP1 isoforms in nuclei affects L1-dependent functions, such as neurite outgrowth and neuronal migration.
Collapse
Affiliation(s)
- Ralf Kleene
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Loers
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gaston Castillo
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
7
|
Increased plasmin-mediated proteolysis of L1CAM in a mouse model of idiopathic normal pressure hydrocephalus. Proc Natl Acad Sci U S A 2021; 118:2010528118. [PMID: 34380733 PMCID: PMC8379912 DOI: 10.1073/pnas.2010528118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is the most common form of adult-onset hydrocephalus, but its etiology is poorly understood. Symptoms develop in previously normal individuals and include gait difficulty, incontinence, and dementia. We recently reported that 15% of iNPH patients harbor heterozygous loss-of-function deletions in CWH43, which encodes a protein that modifies other cell membrane proteins. Mice harboring CWH43 deletions develop hydrocephalus and gait dysfunction. Mutations affecting the L1CAM adhesion protein cause developmental brain abnormalities and hydrocephalus from birth. Here, we show that CWH43 deletion leads to L1CAM hypoglycosylation, decreased L1CAM association with lipid microdomains, increased plasmin-mediated L1CAM cleavage, and decreased L1CAM expression. Thus, decreased L1CAM expression appears to occur in adult-onset iNPH and congenital hydrocephalus. Idiopathic normal pressure hydrocephalus (iNPH) is a common neurological disorder that is characterized by enlarged cerebral ventricles, gait difficulty, incontinence, and dementia. iNPH usually develops after the sixth decade of life in previously asymptomatic individuals. We recently reported that loss-of-function deletions in CWH43 lead to the development of iNPH in a subgroup of patients, but how this occurs is poorly understood. Here, we show that deletions in CWH43 decrease expression of the cell adhesion molecule, L1CAM, in the brains of CWH43 mutant mice and in human HeLa cells harboring a CWH43 deletion. Loss-of-function mutations in L1CAM are a common cause of severe neurodevelopmental defects that include congenital X-linked hydrocephalus. Mechanistically, we find that CWH43 deletion leads to decreased N-glycosylation of L1CAM, decreased association of L1CAM with cell membrane lipid microdomains, increased L1CAM cleavage by plasmin, and increased shedding of cleaved L1CAM in the cerebrospinal fluid. CWH43 deletion also decreased L1CAM nuclear translocation, suggesting decreased L1CAM intracellular signaling. Importantly, the increase in L1CAM cleavage occurred primarily in the ventricular and subventricular zones where brain CWH43 is most highly expressed. Thus, CWH43 deletions may contribute to adult-onset iNPH by selectively downregulating L1CAM in the ventricular and subventricular zone.
Collapse
|
8
|
Desse VE, Blanchette CR, Nadour M, Perrat P, Rivollet L, Khandekar A, Bénard CY. Neuronal post-developmentally acting SAX-7S/L1CAM can function as cleaved fragments to maintain neuronal architecture in C. elegans. Genetics 2021; 218:6296841. [PMID: 34115111 DOI: 10.1093/genetics/iyab086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/24/2021] [Indexed: 01/09/2023] Open
Abstract
Whereas remarkable advances have uncovered mechanisms that drive nervous system assembly, the processes responsible for the lifelong maintenance of nervous system architecture remain poorly understood. Subsequent to its establishment during embryogenesis, neuronal architecture is maintained throughout life in the face of the animal's growth, maturation processes, the addition of new neurons, body movements, and aging. The C. elegans protein SAX-7, homologous to the vertebrate L1 protein family of neural adhesion molecules, is required for maintaining the organization of neuronal ganglia and fascicles after their successful initial embryonic development. To dissect the function of sax-7 in neuronal maintenance, we generated a null allele and sax-7S-isoform-specific alleles. We find that the null sax-7(qv30) is, in some contexts, more severe than previously described mutant alleles, and that the loss of sax-7S largely phenocopies the null, consistent with sax-7S being the key isoform in neuronal maintenance. Using a sfGFP::SAX-7S knock-in, we observe sax-7S to be predominantly expressed across the nervous system, from embryogenesis to adulthood. Yet, its role in maintaining neuronal organization is ensured by post-developmentally acting SAX-7S, as larval transgenic sax-7S(+) expression alone is sufficient to profoundly rescue the null mutants' neuronal maintenance defects. Moreover, the majority of the protein SAX-7 appears to be cleaved, and we show that these cleaved SAX-7S fragments together, not individually, can fully support neuronal maintenance. These findings contribute to our understanding of the role of the conserved protein SAX-7/L1CAM in long-term neuronal maintenance, and may help decipher processes that go awry in some neurodegenerative conditions.
Collapse
Affiliation(s)
- Virginie E Desse
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Cassandra R Blanchette
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Malika Nadour
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Paola Perrat
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lise Rivollet
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
| | - Anagha Khandekar
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Claire Y Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Université du Québec à Montréal, Montréal, QC H2X 1Y4, Canada
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
9
|
Sonnenberg SB, Rauer J, Göhr C, Gorinski N, Schade SK, Abdel Galil D, Naumenko V, Zeug A, Bischoff SC, Ponimaskin E, Guseva D. The 5-HT 4 receptor interacts with adhesion molecule L1 to modulate morphogenic signaling in neurons. J Cell Sci 2021; 134:jcs.249193. [PMID: 33536244 DOI: 10.1242/jcs.249193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/19/2021] [Indexed: 11/20/2022] Open
Abstract
Morphological remodeling of dendritic spines is critically involved in memory formation and depends on adhesion molecules. Serotonin receptors are also implicated in this remodeling, though the underlying mechanisms remain enigmatic. Here, we uncovered a signaling pathway involving the adhesion molecule L1CAM (L1) and serotonin receptor 5-HT4 (5-HT4R, encoded by HTR4). Using Förster resonance energy transfer (FRET) imaging, we demonstrated a physical interaction between 5-HT4R and L1, and found that 5-HT4R-L1 heterodimerization facilitates mitogen-activated protein kinase activation in a Gs-dependent manner. We also found that 5-HT4R-L1-mediated signaling is involved in G13-dependent modulation of cofilin-1 activity. In hippocampal neurons in vitro, the 5-HT4R-L1 pathway triggers maturation of dendritic spines. Thus, the 5-HT4R-L1 signaling module represents a previously unknown molecular pathway regulating synaptic remodeling.
Collapse
Affiliation(s)
| | - Jonah Rauer
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Christoph Göhr
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Nataliya Gorinski
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Sophie Kristin Schade
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Dalia Abdel Galil
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Vladimir Naumenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - André Zeug
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Stephan C Bischoff
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Germany
| | - Evgeni Ponimaskin
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany .,Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Daria Guseva
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany .,Department of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Germany
| |
Collapse
|
10
|
Kleene R, Lutz D, Loers G, Bork U, Borgmeyer U, Hermans-Borgmeyer I, Schachner M. Revisiting the proteolytic processing of cell adhesion molecule L1. J Neurochem 2020; 157:1102-1117. [PMID: 32986867 DOI: 10.1111/jnc.15201] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/18/2020] [Accepted: 09/22/2020] [Indexed: 01/02/2023]
Abstract
The important functions of cell adhesion molecule L1 in the nervous system depend on diverse proteolytic enzymes which generate different L1 fragments. It has been reported that cleavage in the third fibronectin type III (FNIII) homologous domain generates the fragments L1-80 and L1-140, while cleavage in the first FNIII domain yields the fragments L1-70 and L1-135. These results raised questions concerning the L1 cleavage sites. We thus generated gene-edited mice expressing L1 with mutations of the cleavage sites either in the first or third FNIII domain. By immunoprecipitations and immunoblot analyses using brain homogenates and different L1 antibodies, we show that L1-70 and L1-135 are generated in wild-type mice, but not or only to a low extent in L1 mutant mice. L1-80 and L1-140 were not detected in wild-type or mutant mice. Mass spectrometry confirmed the results from immunoprecipitations and immunoblot analyses. Based on these observations, we propose that L1-70 and L1-135 are the predominant fragments in the mouse nervous system and that the third FNIII domain is decisive for generating these fragments. Treatment of cultured cerebellar neurons with trypsin or plasmin, which were both proposed to generate L1-80 and L1-140 by cleaving in the third FNIII domain, showed by immunoprecipitations and immunoblot analyses that both proteases lead to the generation of L1-70 and L1-135, but not L1-80 and L1-140. We discuss previous observations on the basis of our new results and propose a novel view on the molecular features that render previous and present observations compatible.
Collapse
Affiliation(s)
- Ralf Kleene
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - David Lutz
- Institute for Structural Neurobiology, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.,Department for Neuroanatomy and Molecular Brain Research, Ruhr-University Bochum, Bochum, Germany
| | - Gabriele Loers
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ute Bork
- Research Group Biosynthesis of Neural Structures, Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Uwe Borgmeyer
- Scientific Service Group for Transgenic Animals, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Irm Hermans-Borgmeyer
- Scientific Service Group for Transgenic Animals, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
11
|
Girbes Minguez M, Wolters-Eisfeld G, Lutz D, Buck F, Schachner M, Kleene R. The cell adhesion molecule L1 interacts with nuclear proteins via its intracellular domain. FASEB J 2020; 34:9869-9883. [PMID: 32533745 DOI: 10.1096/fj.201902242r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/05/2023]
Abstract
Proteolytic cleavage of the cell adhesion molecule L1 (L1) in brain tissue and in cultured cerebellar neurons results in the generation and nuclear import of a 30 kDa fragment comprising most of L1's C-terminal, intracellular domain. In search of molecules that interact with this domain, we performed affinity chromatography with the recombinant intracellular L1 domain and a nuclear extract from mouse brains, and identified potential nuclear L1 binding partners involved in transcriptional regulation, RNA processing and transport, DNA repair, chromatin remodeling, and nucleocytoplasmic transport. By co-immunoprecipitation and enzyme-linked immunosorbent assay using recombinant proteins, we verified the direct interaction between L1 and the nuclear binding partners non-POU domain containing octamer-binding protein and splicing factor proline/glutamine-rich. The proximity ligation assay confirmed this close interaction in cultures of cerebellar granule cells. Our findings suggest that L1 fragments regulate multiple nuclear functions in the nervous system. We discuss possible physiological and pathological roles of these interactions in regulation of chromatin structure, gene expression, RNA processing, and DNA repair.
Collapse
Affiliation(s)
- Maria Girbes Minguez
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - David Lutz
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Buck
- Zentrum für Diagnostik, Institut für Klinische Chemie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
12
|
Płatek R, Grycz K, Więckowska A, Czarkowska-Bauch J, Skup M. L1 Cell Adhesion Molecule Overexpression Down Regulates Phosphacan and Up Regulates Structural Plasticity-Related Genes Rostral and Caudal to the Complete Spinal Cord Transection. J Neurotrauma 2019; 37:534-554. [PMID: 31426714 DOI: 10.1089/neu.2018.6103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
L1 cell adhesion molecule (L1CAM) supports spinal cord cellular milieu after contusion and compression lesions, contributing to neuroprotection, promoting axonal outgrowth, and reducing outgrowth-inhibitory molecules in lesion proximity. We extended investigations into L1CAM molecular targets and explored long-distance effects of L1CAM rostral and caudal to complete spinal cord transection (SCT) in adult rats. L1CAM overexpression in neurons and glia after Th10/Th11 SCT was achieved using adeno-associated viral vector serotype 5 (AAV5) injected into an L1-lumbar segment immediately after transection. At 5 weeks, a L1CAM mRNA profound decrease detected rostral and caudal to the transection site was alleviated by AAV5-L1CAM treatment, with increased endogenous L1CAM rostral to the SCT. Transected corticospinal tract fibers showed attenuated retraction after treatment, accompanied by a multi-segmental increase of lesion-reduced expression of adenylate cyclase 1 (Adcy1), synaptophysin, growth-associated protein 43, and myelin basic protein genes caudal to transection, and Adcy1 rostral to transection. In parallel, chondroitin sulfate proteoglycan phosphacan elevated after SCT was downregulated after treatment. Low-molecular L1CAM isoforms generated after spinalization indicated the involvement of sheddases in L1CAM processing and long-distance effects. A disintegrin and metalloproteinase (ADAM)10 sheddase immunoreactivity, stronger in AAV5-L1CAM than AAV5- enhanced green fluorescent protein (EGFP)-transduced motoneurons indicated local ADAM10 upregulation by L1CAM. The results suggest that increased L1CAM availability and penetration of diffusible L1CAM fragments post-lesion induce both local and long-distance neuronal and glial responses toward better neuronal maintenance, neurite growth, and myelination. Despite the fact that intervention promoted beneficial molecular changes, kinematic analysis of hindlimb movements showed minor improvement, indicating that spinalized rats require longer L1CAM treatment to regain locomotor functions.
Collapse
Affiliation(s)
- Rafał Płatek
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Kamil Grycz
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | |
Collapse
|
13
|
Linneberg C, Toft CLF, Kjaer-Sorensen K, Laursen LS. L1cam-mediated developmental processes of the nervous system are differentially regulated by proteolytic processing. Sci Rep 2019; 9:3716. [PMID: 30842511 PMCID: PMC6403279 DOI: 10.1038/s41598-019-39884-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/08/2018] [Indexed: 11/09/2022] Open
Abstract
Normal brain development depends on tight temporal and spatial regulation of connections between cells. Mutations in L1cam, a member of the immunoglobulin (Ig) superfamily that mediate cell-cell contacts through homo- and heterophilic interactions, are associated with several developmental abnormalities of the nervous system, including mental retardation, limb spasticity, hydrocephalus, and corpus callosum aplasia. L1cam has been reported to be shed from the cell surface, but the significance of this during different phases of brain development is unknown. We here show that ADAM10-mediated shedding of L1cam is regulated by its fibronectin type III (FNIII) domains. Specifically, the third FNIII domain is important for maintaining a conformation where access to a membrane proximal cleavage site is restricted. To define the role of ADAM10/17/BACE1-mediated shedding of L1cam during brain development, we used a zebrafish model system. Knockdown of the zebrafish, l1camb, caused hydrocephalus, defects in axonal outgrowth, and myelination abnormalities. Rescue experiments with proteinase-resistant and soluble L1cam variants showed that proteolytic cleavage is not required for normal axonal outgrowth and development of the ventricular system. In contrast, metalloproteinase-mediated shedding is required for efficient myelination, and only specific fragments are able to mediate this stimulatory function of the shedded L1cam.
Collapse
Affiliation(s)
- Cecilie Linneberg
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark
| | - Christian Liebst Frisk Toft
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark
| | - Lisbeth S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000, Aarhus C, Denmark.
| |
Collapse
|
14
|
Wu JD, Hong CQ, Huang WH, Wei XL, Zhang F, Zhuang YX, Zhang YQ, Zhang GJ. L1 Cell Adhesion Molecule and Its Soluble Form sL1 Exhibit Poor Prognosis in Primary Breast Cancer Patients. Clin Breast Cancer 2018; 18:e851-e861. [PMID: 29510897 DOI: 10.1016/j.clbc.2017.12.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The L1 cell adhesion molecule (L1-CAM) and its soluble form sL1 play a prominent role in invasion and metastasis in several cancers. However, its association with breast cancer is still unclear. PATIENTS AND METHODS We analyzed L1-CAM expression and serum sL1 levels in cancer and para-carcinoma tissues from 162 consecutive patients with primary invasive breast cancer (PBC) using immunohistochemistry and an enzyme-linked immunosorbent assay, respectively. The serum sL1 levels were also examined in 38 patients with benign breast disease and 36 healthy controls. RESULTS L1-CAM was expressed more frequently in cancer tissues than in para-carcinoma tissues (24.1% vs. 5.6%; P < .001), and the mean sL1 levels were significantly greater in PBC than in those with benign breast disease and healthy controls (P = .027). Both L1-CAM+ expression and higher mean sL1 levels correlated significantly with larger tumor size, lymph node involvement, higher histologic grade, advanced TNM stage, and shorter disease-free survival for PBC patients. Moreover, higher mean sL1 levels were also significantly associated with estrogen receptor-α-negative expression, human epidermal growth factor receptor 2-positive (HER2+) expression, HER2-enriched and triple-negative molecular subtypes, and L1-CAM+ expression (P < .05). On multivariate analysis, larger tumor size, nodal involvement, HER2+, and higher sL1 levels (≥ 0.7 ng/mL) were independent factors associated with L1-CAM+ expression (P < .05). No association was found between L1-CAM expression or sL1 level with age, gender, histologic type, or expression of progesterone receptor, Ki-67, p53, or vascular endothelial growth factor C (P > .05). CONCLUSION These results indicate that L1-CAM and sL1 are elevated in PBC and both might affect the prognosis of PBC patients. In addition, sL1 might be a useful marker for screening and diagnosis.
Collapse
Affiliation(s)
- Jun-Dong Wu
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Chao-Qun Hong
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Wen-He Huang
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Fan Zhang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Yi-Xuan Zhuang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Yong-Qu Zhang
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Guo-Jun Zhang
- Changjiang Scholar's Laboratory, Shantou University Medical College, Guangdong, China.
| |
Collapse
|
15
|
Kraus K, Kleene R, Henis M, Braren I, Kataria H, Sharaf A, Loers G, Schachner M, Lutz D. A Fragment of Adhesion Molecule L1 Binds to Nuclear Receptors to Regulate Synaptic Plasticity and Motor Coordination. Mol Neurobiol 2018; 55:7164-7178. [PMID: 29383692 DOI: 10.1007/s12035-018-0901-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/10/2018] [Indexed: 02/05/2023]
Abstract
Proteolytic cleavage of the neuronal isoform of the murine cell adhesion molecule L1, triggered by stimulation of the cognate L1-dependent signaling pathways, results in the generation and nuclear import of an L1 fragment that contains the intracellular domain, the transmembrane domain, and part of the extracellular domain. Here, we show that the LXXLL and FXXLF motifs in the extracellular and transmembrane domain of this L1 fragment mediate the interaction with the nuclear estrogen receptors α (ERα) and β (ERβ), peroxisome proliferator-activated receptor γ (PPARγ), and retinoid X receptor β (RXRβ). Mutations of the LXXLL motif in the transmembrane domain and of the FXXLF motif in the extracellular domain disturb the interaction of the L1 fragment with these nuclear receptors and, when introduced by viral transduction into mouse embryos in utero, result in impaired motor coordination, learning and memory, as well as synaptic connectivity in the cerebellum, in adulthood. These impairments are similar to those observed in the L1-deficient mouse. Our findings suggest that the interplay of nuclear L1 and distinct nuclear receptors is associated with synaptic contact formation and plasticity.
Collapse
Affiliation(s)
- Kristina Kraus
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melad Henis
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Anatomy and Histology, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ingke Braren
- Vector Core Unit, Institut für Experimentelle Pharmakologie und Toxikologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ahmed Sharaf
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Gabriele Loers
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
| | - David Lutz
- Arbeitsgruppe für Biosynthese Neuraler Strukturen, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
- Institut für Strukturelle Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| |
Collapse
|
16
|
Lutz D, Sharaf A, Drexler D, Kataria H, Wolters-Eisfeld G, Brunne B, Kleene R, Loers G, Frotscher M, Schachner M. Proteolytic cleavage of transmembrane cell adhesion molecule L1 by extracellular matrix molecule Reelin is important for mouse brain development. Sci Rep 2017; 7:15268. [PMID: 29127326 PMCID: PMC5681625 DOI: 10.1038/s41598-017-15311-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/25/2017] [Indexed: 02/05/2023] Open
Abstract
The cell adhesion molecule L1 and the extracellular matrix protein Reelin play crucial roles in the developing nervous system. Reelin is known to activate signalling cascades regulating neuronal migration by binding to lipoprotein receptors. However, the interaction of Reelin with adhesion molecules, such as L1, has remained poorly explored. Here, we report that full-length Reelin and its N-terminal fragments N-R2 and N-R6 bind to L1 and that full-length Reelin and its N-terminal fragment N-R6 proteolytically cleave L1 to generate an L1 fragment with a molecular mass of 80 kDa (L1-80). Expression of N-R6 and generation of L1-80 coincide in time at early developmental stages of the cerebral cortex. Reelin-mediated generation of L1-80 is involved in neurite outgrowth and in stimulation of migration of cultured cortical and cerebellar neurons. Morphological abnormalities in layer formation of the cerebral cortex of L1-deficient mice partially overlap with those of Reelin-deficient reeler mice. In utero electroporation of L1-80 into reeler embryos normalised the migration of cortical neurons in reeler embryos. The combined results indicate that the direct interaction between L1 and Reelin as well as the Reelin-mediated generation of L1-80 contribute to brain development at early developmental stages.
Collapse
Affiliation(s)
- David Lutz
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany. .,Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Ahmed Sharaf
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Dagmar Drexler
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Bianka Brunne
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gabriele Loers
- Institute for Biosynthesis of Neural Structures, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA. .,Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guandong, 515041, China.
| |
Collapse
|
17
|
Lutz D, Kataria H, Kleene R, Loers G, Chaudhary H, Guseva D, Wu B, Jakovcevski I, Schachner M. Myelin Basic Protein Cleaves Cell Adhesion Molecule L1 and Improves Regeneration After Injury. Mol Neurobiol 2016; 53:3360-3376. [PMID: 26081148 DOI: 10.1007/s12035-015-9277-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/01/2015] [Indexed: 02/05/2023]
Abstract
Myelin basic protein (MBP) is a serine protease that cleaves neural cell adhesion molecule L1 and generates a transmembrane L1 fragment which facilitates L1-dependent functions in vitro, such as neurite outgrowth, neuronal cell migration and survival, myelination by Schwann cells as well as Schwann cell proliferation, migration, and process formation. Ablation and blocking of MBP or disruption of its proteolytic activity by mutation of a proteolytically active serine residue abolish L1-dependent cellular responses. In utero injection of adeno-associated virus encoding proteolytically active MBP into MBP-deficient shiverer mice normalizes differentiation, myelination, and synaptogenesis in the developing postnatal spinal cord, in contrast to proteolytically inactive MBP. Application of active MBP to the injured wild-type spinal cord and femoral nerve augments levels of a transmembrane L1 fragment, promotes remyelination, and improves functional recovery after injury. Application of MBP antibody impairs recovery. Virus-mediated expression of active MBP in the lesion site after spinal cord injury results in improved functional recovery, whereas injection of virus encoding proteolytically inactive MBP fails to do so. The present study provides evidence for a novel L1-mediated function of MBP in the developing spinal cord and in the injured adult mammalian nervous system that leads to enhanced recovery after acute trauma.
Collapse
Affiliation(s)
- David Lutz
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Hardeep Kataria
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Harshita Chaudhary
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daria Guseva
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Cellular Neurobiology, Medical School Hannover, Hannover, Germany
| | - Bin Wu
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Melitta Schachner
- Melitta Schachner, Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, Guangdong, 515041, China.
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
18
|
Lee HD, Kim YH, Koo BH, Kim DS. The ADAM15 ectodomain is shed from secretory exosomes. BMB Rep 2016; 48:277-82. [PMID: 25208722 PMCID: PMC4578567 DOI: 10.5483/bmbrep.2015.48.5.161] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Indexed: 12/27/2022] Open
Abstract
We demonstrated previously that a disintegrin and metalloproteinase 15 (ADAM15) is released into the extracellular space as an exosomal component, and that ADAM15-rich exosomes have tumor suppressive functions. However, the suppressive mechanism of ADAM15-rich exosomes remains unclear. In this study, we show that the ADAM15 ectodomain is cleaved from released exosomes. This shedding process of the ADAM15 ectodomain was dramatically enhanced in conditioned ovarian cancer cell medium. Proteolytic cleavage was completely blocked by phenylmethylsulfonyl fluoride, indicating that a serine protease is responsible for exosomal ADAM15 shedding. Experimental evidence indicates that the ADAM15 ectodomain itself has comparable functions with those of ADAM15-rich exosomes, which effectively inhibit vitronectininduced cancer cell migration and activation of the MEK/extracellular regulated kinase signaling pathway. We present a tumor suppressive mechanism for ADAM15 exosomes and provide insight into the functional significance of exosomes that generate tumor-inhibitory factors. [BMB Reports 2015; 48(5): 277-282]
Collapse
Affiliation(s)
- Hee Doo Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Yeon Hyang Kim
- Department of Bioinformatics, Korea Polytechnics, Nonsan 320-905, Korea
| | - Bon-Hun Koo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Doo-Sik Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
19
|
Lutz D, Wolters-Eisfeld G, Schachner M, Kleene R. Cathepsin E generates a sumoylated intracellular fragment of the cell adhesion molecule L1 to promote neuronal and Schwann cell migration as well as myelination. J Neurochem 2014; 128:713-24. [DOI: 10.1111/jnc.12473] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 09/11/2013] [Accepted: 09/27/2013] [Indexed: 02/05/2023]
Affiliation(s)
- David Lutz
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Gerrit Wolters-Eisfeld
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience; Rutgers University; Piscataway New Jersey USA
- Center for Neuroscience; Shantou University Medical College; Shantou China
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie; Universitätsklinikum Hamburg-Eppendorf; Hamburg Germany
| |
Collapse
|
20
|
Valiente M, Obenauf AC, Jin X, Chen Q, Zhang XHF, Lee DJ, Chaft JE, Kris MG, Huse JT, Brogi E, Massagué J. Serpins promote cancer cell survival and vascular co-option in brain metastasis. Cell 2014; 156:1002-16. [PMID: 24581498 PMCID: PMC3988473 DOI: 10.1016/j.cell.2014.01.040] [Citation(s) in RCA: 634] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 09/16/2013] [Accepted: 01/14/2014] [Indexed: 12/24/2022]
Abstract
Brain metastasis is an ominous complication of cancer, yet most cancer cells that infiltrate the brain die of unknown causes. Here, we identify plasmin from the reactive brain stroma as a defense against metastatic invasion, and plasminogen activator (PA) inhibitory serpins in cancer cells as a shield against this defense. Plasmin suppresses brain metastasis in two ways: by converting membrane-bound astrocytic FasL into a paracrine death signal for cancer cells, and by inactivating the axon pathfinding molecule L1CAM, which metastatic cells express for spreading along brain capillaries and for metastatic outgrowth. Brain metastatic cells from lung cancer and breast cancer express high levels of anti-PA serpins, including neuroserpin and serpin B2, to prevent plasmin generation and its metastasis-suppressive effects. By protecting cancer cells from death signals and fostering vascular co-option, anti-PA serpins provide a unifying mechanism for the initiation of brain metastasis in lung and breast cancers.
Collapse
Affiliation(s)
- Manuel Valiente
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna C Obenauf
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xin Jin
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qing Chen
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xiang H-F Zhang
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Derek J Lee
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jamie E Chaft
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mark G Kris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason T Huse
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Metastasis Research Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Howard Hughes Medical Institute, Chevy Chase, MD 21205, USA.
| |
Collapse
|
21
|
Differential effects of human L1CAM mutations on complementing guidance and synaptic defects in Drosophila melanogaster. PLoS One 2013; 8:e76974. [PMID: 24155914 PMCID: PMC3796554 DOI: 10.1371/journal.pone.0076974] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/05/2013] [Indexed: 01/17/2023] Open
Abstract
A large number of different pathological L1CAM mutations have been identified that result in a broad spectrum of neurological and non-neurological phenotypes. While many of these mutations have been characterized for their effects on homophilic and heterophilic interactions, as well as expression levels in vitro, there are only few studies on their biological consequences in vivo. The single L1-type CAM gene in Drosophila, neuroglian (nrg), has distinct functions during axon guidance and synapse formation and the phenotypes of nrg mutants can be rescued by the expression of human L1CAM. We previously showed that the highly conserved intracellular FIGQY Ankyrin-binding motif is required for L1CAM-mediated synapse formation, but not for neurite outgrowth or axon guidance of the Drosophila giant fiber (GF) neuron. Here, we use the GF as a model neuron to characterize the pathogenic L120V, Y1070C, C264Y, H210Q, E309K and R184Q extracellular L1CAM missense mutations and a L1CAM protein with a disrupted ezrin-moesin-radixin (ERM) binding site to investigate the signaling requirements for neuronal development. We report that different L1CAM mutations have distinct effects on axon guidance and synapse formation. Furthermore, L1CAM homophilic binding and signaling via the ERM motif is essential for axon guidance in Drosophila. In addition, the human pathological H210Q, R184Q and Y1070C, but not the E309K and L120V L1CAM mutations affect outside-in signaling via the FIGQY Ankyrin binding domain which is required for synapse formation. Thus, the pathological phenotypes observed in humans are likely to be caused by the disruption of signaling required for both, guidance and synaptogenesis.
Collapse
|
22
|
Kiefel H, Bondong S, Hazin J, Ridinger J, Schirmer U, Riedle S, Altevogt P. L1CAM: a major driver for tumor cell invasion and motility. Cell Adh Migr 2012; 6:374-84. [PMID: 22796939 DOI: 10.4161/cam.20832] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The L1 cell adhesion molecule (L1CAM) plays a major role in the development of the nervous system and in the malignancy of human tumors. In terms of biological function, L1CAM comes along in two different flavors: (1) a static function as a cell adhesion molecule that acts as a glue between cells; (2) a motility promoting function that drives cell migration during neural development and supports metastasis of human cancers. Important factors that contribute to the switch in the functional mode of L1CAM are: (1) the cleavage from the cell surface by membrane proximal proteolysis and (2) the ability to change binding partners and engage in L1CAM-integrin binding. Recent studies have shown that the cleavage of L1CAM by metalloproteinases and the binding of L1CAM to integrins via its RGD-motif in the sixth Ig-domain activate signaling pathways distinct from the ones elicited by homophilic binding. Here we highlight important features of L1CAM proteolysis and the signaling of L1CAM via integrin engagement. The novel insights into L1CAM downstream signaling and its regulation during tumor progression and epithelial-mesenchymal transition (EMT) will lead to a better understanding of the dualistic role of L1CAM as a cell adhesion and/or motility promoting cell surface molecule.
Collapse
Affiliation(s)
- Helena Kiefel
- Translational Immunology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
23
|
Lutz D, Wolters-Eisfeld G, Joshi G, Djogo N, Jakovcevski I, Schachner M, Kleene R. Generation and nuclear translocation of sumoylated transmembrane fragment of cell adhesion molecule L1. J Biol Chem 2012; 287:17161-17175. [PMID: 22431726 DOI: 10.1074/jbc.m112.346759] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The functions of the cell adhesion molecule L1 in the developing and adult nervous system are triggered by homophilic and heterophilic interactions that stimulate signal transductions that activate cellular responses. Here, we show that stimulation of signaling by function-triggering L1 antibodies or L1-Fc leads to serine protease-dependent cleavage of full-length L1 at the plasma membrane and generation of a sumoylated transmembrane 70-kDa fragment comprising the intracellular and transmembrane domains and part of the extracellular domain. The 70-kDa transmembrane fragment is transported from the plasma membrane to a late endosomal compartment, released from endosomal membranes into the cytoplasm, and transferred from there into the nucleus by a pathway that depends on importin and chromatin-modifying protein 1. Mutation of the sumoylation site at Lys(1172) or of the nuclear localization signal at Lys(1147) abolished L1-stimulated generation or nuclear import of the 70-kDa fragment, respectively. Nuclear import of the 70-kDa fragment may activate cellular responses in parallel or in association with phosphorylation-dependent signaling pathways. Alterations in the levels of the 70-kDa fragment during development and in the adult after spinal cord injury or in a mouse model of Alzheimer disease suggest that this fragment is functionally implicated in development, regeneration, neurodegeneration, tumorigenesis, and possibly synaptic plasticity in the mature nervous system.
Collapse
Affiliation(s)
- David Lutz
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gunjan Joshi
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nevena Djogo
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Igor Jakovcevski
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Melitta Schachner
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, China.
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
24
|
Chen MM, Lee CY, Leland HA, Silletti S. Modification of the L1-CAM carboxy-terminus in pancreatic adenocarcinoma cells. Tumour Biol 2010; 32:347-57. [PMID: 21080252 PMCID: PMC3041914 DOI: 10.1007/s13277-010-0127-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Accepted: 10/29/2010] [Indexed: 01/08/2023] Open
Abstract
The neural cell adhesion molecule L1 has recently been shown to be expressed in pancreatic adenocarcinoma (PDAC) cells. In this report, we demonstrate that L1 is expressed by moderately- to poorly-differentiated PDAC cells in situ, and that L1 expression is a predictor of poor patient survival. In vitro, reduced reactivity of an anti-L1 carboxy-terminus-specific antibody was observed in the more poorly differentiated fast-growing (FG) variant of the COLO357 population, versus its well-differentiated slow-growing (SG) counterpart, even though they express equivalent total L1. The carboxy-terminus of L1 mediates binding to the MAP kinase-regulating protein RanBPM and mutation of T1247/S1248 within this region attenuates the expression of malignancy associated proteins and L1-induced tumorigenicity in mice. Therefore, we reasoned that the differential epitope exposure observed might be indicative of modifications responsible for regulating these events. However, epitope mapping demonstrated that the major determinant of binding was actually N1251; mutation of T1247 and S1248, alone or together, had little effect on C20 binding. Moreover, cluster assays using CD25 ectodomain/L1 cytoplasmic domain chimeras demonstrated the N1251-dependent, RanBPM-independent stimulation of erk phosphorylation in these cells. Reactivity of this antibody also reflects the differential exposure of extracellular epitopes in these COLO357 sublines, consistent with the previous demonstration of L1 ectodomain conformation modulation by intracellular modifications. These data further support a central role for L1 in PDAC, and define a specific role for carboxy-terminal residues including N1251 in the regulation of L1 activity in PDAC cells.
Collapse
Affiliation(s)
- Maxine M Chen
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
25
|
Schäfer MKE, Altevogt P. L1CAM malfunction in the nervous system and human carcinomas. Cell Mol Life Sci 2010; 67:2425-37. [PMID: 20237819 PMCID: PMC11115577 DOI: 10.1007/s00018-010-0339-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 01/30/2010] [Accepted: 02/11/2010] [Indexed: 12/14/2022]
Abstract
Research over the last 25 years on the cell adhesion molecule L1 has revealed its pivotal role in nervous system function. Mutations of the human L1CAM gene have been shown to cause neurodevelopmental disorders such as X-linked hydrocephalus, spastic paraplegia and mental retardation. Impaired L1 function has been also implicated in the aetiology of fetal alcohol spectrum disorders, defective enteric nervous system development and malformations of the renal system. Importantly, aberrant expression of L1 has emerged as a critical factor in the development of human carcinomas, where it enhances cell proliferation, motility and chemoresistance. This discovery promoted collaborative work between tumour biologists and neurobiologists, which has led to a substantial expansion of the basic knowledge about L1 function and regulation. Here we provide an overview of the pathological conditions caused by L1 malfunction. We further discuss how the available data on gene regulation, molecular interactions and posttranslational processing of L1 may contribute to a better understanding of associated neurological and cancerous diseases.
Collapse
Affiliation(s)
- Michael K E Schäfer
- Center for Neurosciences, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
26
|
Min JK, Kim JM, Li S, Lee JW, Yoon H, Ryu CJ, Jeon SH, Lee JH, Kim JY, Yoon HK, Lee YK, Kim BH, Son YS, Choi HS, Lim NK, Kim DG, Hong HJ. L1 Cell Adhesion Molecule Is a Novel Therapeutic Target in Intrahepatic Cholangiocarcinoma. Clin Cancer Res 2010; 16:3571-80. [DOI: 10.1158/1078-0432.ccr-09-3075] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
27
|
Abstract
The L1 family of cell adhesion molecules (L1CAMs) in vertebrates has long been studied for its roles in nervous system development and function. Members of this family have been associated with distinct neurological disorders that include CRASH, autism, 3p syndrome, and schizophrenia. The conservation of L1CAMs in Drosophila and Caenorhabditis elegans allows the opportunity to take advantage of these simple model organisms and their accessible genetic manipulations to dissect L1CAM functions and mechanisms of action. This review summarizes the discoveries of L1CAMs made in C. elegans, showcasing this simple model organism as a powerful system to uncover L1CAM mechanisms and roles in healthy and diseased states.
Collapse
Affiliation(s)
- Lihsia Chen
- Department of Genetics, Cell Biology, and Development, Developmental Biology Center, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
28
|
Chen MM, Lee CY, Leland HA, Lin GY, Montgomery AM, Silletti S. Inside-out regulation of L1 conformation, integrin binding, proteolysis, and concomitant cell migration. Mol Biol Cell 2010; 21:1671-85. [PMID: 20335502 PMCID: PMC2869374 DOI: 10.1091/mbc.e09-10-0900] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The ectodomain structure and function of the neural cell adhesion molecule L1 is shown to be regulated by the intracellular phosphorylation of a novel threonine, T1172. In pancreatic cancer cells, T1172 exhibits steady-state saturated phosphorylation, an event regulated by CKII and PKC, and which further regulates cell migration. Previous reports on the expression of the cell adhesion molecule L1 in pancreatic ductal adenocarcinoma (PDAC) cells range from absent to high. Our data demonstrate that L1 is expressed in poorly differentiated PDAC cells in situ and that threonine-1172 (T1172) in the L1 cytoplasmic domain exhibits steady-state saturated phosphorylation in PDAC cells in vitro and in situ. In vitro studies support roles for casein kinase II and PKC in this modification, consistent with our prior studies using recombinant proteins. Importantly, T1172 phosphorylation drives, or is associated with, a change in the extracellular structure of L1, consistent with a potential role in regulating the shift between the closed conformation and the open, multimerized conformation of L1. We further demonstrate that these distinct conformations exhibit differential binding to integrins αvβ3 and αvβ5 and that T1172 regulates cell migration in a matrix-specific manner and is required for a disintegrin and metalloproteinase-mediated shedding of the L1 ectodomain that has been shown to regulate cell migration. These data define a specific role for T1172 of L1 in regulating aspects of pancreatic adenocarcinoma cell phenotype and suggest the need for further studies to elucidate the specific ramifications of L1 expression and T1172 phosphorylation in the pathobiology of pancreatic cancer.
Collapse
Affiliation(s)
- Maxine M Chen
- Moores Cancer Center and Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
29
|
van Kilsdonk JWJ, van Kempen LCLT, van Muijen GNP, Ruiter DJ, Swart GWM. Soluble adhesion molecules in human cancers: sources and fates. Eur J Cell Biol 2010; 89:415-27. [PMID: 20227133 DOI: 10.1016/j.ejcb.2009.11.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 11/20/2009] [Indexed: 12/18/2022] Open
Abstract
Adhesion molecules endow tumor cells with the necessary cell-cell contacts and cell-matrix interactions. As such, adhesion molecules are involved in cell signalling, proliferation and tumor growth. Rearrangements in the adhesion repertoire allow tumor cells to migrate, invade and form metastases. Besides these membrane-bound adhesion molecules several soluble adhesion molecules are detected in the supernatant of tumor cell lines and patient body fluids. Truncated soluble adhesion molecules can be generated by several conventional mechanisms, including alternative splicing of mRNA transcripts, chromosomal translocation, and extracellular proteolytic ectodomain shedding. Secretion of vesicles (ectosomes and exosomes) is an alternative mechanism mediating the release of full-length adhesion molecules. Soluble adhesion molecules function as modulators of cell adhesion, induce proteolytic activity and facilitate cell signalling. Additionally, adhesion molecules present on secreted vesicles might be involved in the vesicle-target cell interaction. Based on currently available data, released soluble adhesion molecules contribute to cancer progression and therefore should not be regarded as unrelated and non-functional side products of tumor progression.
Collapse
Affiliation(s)
- Jeroen W J van Kilsdonk
- Department of Biomolecular Chemistry, IMM & NCMLS, Faculty of Science, Radboud University Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | |
Collapse
|
30
|
Guan H, Maness PF. Perisomatic GABAergic innervation in prefrontal cortex is regulated by ankyrin interaction with the L1 cell adhesion molecule. Cereb Cortex 2010; 20:2684-93. [PMID: 20156840 DOI: 10.1093/cercor/bhq016] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The L1 adhesion molecule functions in axon growth and guidance, but a role in synaptic development of cortical inhibitory interneurons is largely unexplored. L1 mediates adhesion by engaging the actin cytoskeleton through binding the actin/spectrin adapter protein ankyrin. Loss of L1-ankyrin interaction impaired process elaboration/branching by GABAergic interneurons, including basket cells, and reduced the number of perisomatic synapses in the cingulate cortex as shown in L1 mutant mice (L1YH) with a mutation in the ankyrin-binding site, either alone or intercrossed with GAD67-enhanced green fluorescence protein reporter mice. Electron microscopy revealed that perisomatic inhibitory synapses but not excitatory synapses in the neuropil were specifically affected. In wild-type cingulate cortex, L1 colocalized with perisomatic synaptic markers, whereas L1 phosphorylation on Tyr(1229) decreased postnatally, correlating with increased ankyrin binding and synaptic development. These results suggest a novel role for L1 engagement with the actin cytoskeleton in development of inhibitory connectivity within the cingulate cortex.
Collapse
Affiliation(s)
- Hanjun Guan
- Department of Biochemistry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
31
|
Yang M, Adla S, Temburni MK, Patel VP, Lagow EL, Brady OA, Tian J, Boulos MI, Galileo DS. Stimulation of glioma cell motility by expression, proteolysis, and release of the L1 neural cell recognition molecule. Cancer Cell Int 2009; 9:27. [PMID: 19874583 PMCID: PMC2776596 DOI: 10.1186/1475-2867-9-27] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 10/29/2009] [Indexed: 12/31/2022] Open
Abstract
Background Malignant glioma cells are particularly motile and can travel diffusely through the brain parenchyma, apparently without following anatomical structures to guide their migration. The neural adhesion/recognition protein L1 (L1CAM; CD171) has been implicated in contributing to stimulation of motility and metastasis of several non-neural cancer types. We explored the expression and function of L1 protein as a stimulator of glioma cell motility using human high-grade glioma surgical specimens and established rat and human glioma cell lines. Results L1 protein expression was found in 17 out of 18 human high-grade glioma surgical specimens by western blotting. L1 mRNA was found to be present in human U-87/LacZ and rat C6 and 9L glioma cell lines. The glioma cell lines were negative for surface full length L1 by flow cytometry and high resolution immunocytochemistry of live cells. However, fixed and permeablized cells exhibited positive staining as numerous intracellular puncta. Western blots of cell line extracts revealed L1 proteolysis into a large soluble ectodomain (~180 kDa) and a smaller transmembrane proteolytic fragment (~32 kDa). Exosomal vesicles released by the glioma cell lines were purified and contained both full-length L1 and the proteolyzed transmembrane fragment. Glioma cell lines expressed L1-binding αvβ5 integrin cell surface receptors. Quantitative time-lapse analyses showed that motility was reduced significantly in glioma cell lines by 1) infection with an antisense-L1 retroviral vector and 2) L1 ectodomain-binding antibodies. Conclusion Our novel results support a model of autocrine/paracrine stimulation of cell motility in glioma cells by a cleaved L1 ectodomain and/or released exosomal vesicles containing L1. This mechanism could explain the diffuse migratory behavior of high-grade glioma cancer cells within the brain.
Collapse
Affiliation(s)
- Muhua Yang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lühmann T, Hänseler P, Grant B, Hall H. The induction of cell alignment by covalently immobilized gradients of the 6th Ig-like domain of cell adhesion molecule L1 in 3D-fibrin matrices. Biomaterials 2009; 30:4503-12. [DOI: 10.1016/j.biomaterials.2009.05.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 05/18/2009] [Indexed: 11/30/2022]
|
33
|
Synaptic plasticity-associated proteases and protease inhibitors in the brain linked to the processing of extracellular matrix and cell adhesion molecules. ACTA ACUST UNITED AC 2009; 4:223-34. [DOI: 10.1017/s1740925x09990172] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Research on the molecular and cellular basis of learning and memory has focused on the mechanisms that underlie the induction and expression of synaptic plasticity. There is increasing evidence that structural changes at the synapse are associated with synaptic plasticity and that extracellular matrix (ECM) components and cell adhesion molecules are associated with these changes. The functions of both groups of molecules can be regulated by proteolysis. In this article we review the roles of selected proteases and protease inhibitors in perisynaptic proteolysis of the ECM and synaptic adhesion proteins and the impact of proteolysis on synaptic modification and cognitive function.
Collapse
|
34
|
Raveh S, Gavert N, Ben-Ze'ev A. L1 cell adhesion molecule (L1CAM) in invasive tumors. Cancer Lett 2009; 282:137-45. [PMID: 19144458 DOI: 10.1016/j.canlet.2008.12.021] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/08/2008] [Accepted: 12/11/2008] [Indexed: 01/11/2023]
Abstract
The L1 cell adhesion molecule (L1CAM) belongs to the immunoglobulin superfamily and was originally identified in the nervous system. Recent studies demonstrated L1CAM expression in various types of cancer, predominantly at the invasive front of tumors and in metastases, suggesting its involvement in advanced stages of tumor progression. Overexpression of L1CAM in normal and cancer cells increased motility, enhanced growth rate and promoted cell transformation and tumorigenicity. Moreover, the expression of L1CAM in tumor cells conferred the capacity to form metastases. These properties of L1CAM, in addition to its cell surface localization, make it a potentially useful diagnostic marker for cancer progression and a candidate for anti-cancer therapy. We review the role of L1CAM in cancer progression with particular emphasis on colon cancer, and the potential of anti-L1CAM antibodies as a therapeutic tool for cancer.
Collapse
Affiliation(s)
- Shani Raveh
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | | | | |
Collapse
|
35
|
Gómez M, Hernández ML, Pazos MR, Tolón RM, Romero J, Fernández-Ruiz J. Colocalization of CB1 receptors with L1 and GAP-43 in forebrain white matter regions during fetal rat brain development: evidence for a role of these receptors in axonal growth and guidance. Neuroscience 2008; 153:687-99. [PMID: 18400407 DOI: 10.1016/j.neuroscience.2008.02.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 01/19/2008] [Accepted: 02/05/2008] [Indexed: 12/13/2022]
Abstract
There is recent evidence supporting the notion that the cannabinoid signaling system plays a modulatory role in the regulation of cell proliferation and migration, survival of neural progenitors, neuritic elongation and guidance, and synaptogenesis. This assumption is based on the fact that cannabinoid 1-type receptors (CB(1) receptors) and their ligands emerge early in brain development and are abundantly expressed in certain brain regions that play key roles in these processes. We have recently presented in vivo evidence showing that this modulatory action might be exerted through regulating the synthesis of the cell adhesion molecule L1 that is also a key element for those processes. To further explore this issue, we conducted here immunohistochemical studies aimed at determining the cellular substrates of CB(1) receptor-L1 interactions in the rat brain during late fetal development. In this period, we previously found that the activation of CB(1) receptors increased L1 synthesis in several forebrain white matter regions but not in gray matter areas. Using double labeling studies, we observed here colocalization of both proteins in fiber tracts including the corpus callosum, the adjacent subcortical white matter, the internal capsule and the anterior commissure. Experiments conducted with cultures of fetal rat cortical nerve cells revealed that L1 is present mainly in neurons but not in glial cells. This fact, together with the results obtained in the double labeling studies, would indicate that L1 and CB(1) receptors should possibly be present in axons elongating through these white matter tracts, or, alternatively, in migrating neurons. Further experiments confirmed the presence of CB(1) receptors in elongating axons, since these receptors colocalized with growth-associated protein 43 (GAP-43), a marker of growth cones, but not with synaptophysin, a marker of active synaptic terminals, in the same forebrain white matter regions. Lastly, using cultured fetal rat cortical neurons, we also observed that the activation of cannabinoid receptors increased the levels of the full-length L1 and altered those of some active proteolytic fragments of this protein whose generation has been associated with specific steps in the process of neuritic elongation in cultured neurons. In summary, we have demonstrated that the effects caused by cannabinoid agonists on L1 are facilitated by the colocalization of this cell adhesion molecule with CB(1) receptors in several forebrain white matter regions during fetal brain development. We have provided strong evidence that this phenomenon occurs in axons elongating through these white matter tracts, and we have explored in vitro how cannabinoid receptors influence L1 levels. Considering the role played by L1 in different events related to neural development, our observations support the occurrence of a physiological mechanism by which the cannabinoid system might regulate the process of axonal growth and guidance through regulating the synthesis and function of L1.
Collapse
Affiliation(s)
- M Gómez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Baculovirus expression and bioactivity of a soluble 140kDa extracellular cleavage fragment of L1 neural cell adhesion molecule. Protein Expr Purif 2008; 57:172-9. [DOI: 10.1016/j.pep.2007.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 10/08/2007] [Accepted: 10/11/2007] [Indexed: 11/21/2022]
|
37
|
Functional dissection of the C. elegans cell adhesion molecule SAX-7, a homologue of human L1. Mol Cell Neurosci 2008; 37:56-68. [DOI: 10.1016/j.mcn.2007.08.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2007] [Revised: 08/08/2007] [Accepted: 08/21/2007] [Indexed: 11/24/2022] Open
|
38
|
Lim SD, Stallcup W, Lefkove B, Govindarajan B, Au KS, Northrup H, Lang D, Fisher DE, Patel A, Amin MB, Arbiser JL. Expression of the neural stem cell markers NG2 and L1 in human angiomyolipoma: are angiomyolipomas neoplasms of stem cells? Mol Med 2007. [PMID: 17592550 DOI: 10.2119/2006-00070.lim] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiomyolipomas are benign tumors of the kidney which express phenotypes of smooth muscle, fat, and melanocytes. These tumors appear with increased frequency in the autosomal dominant disorder tuberous sclerosis and are the leading cause of morbidity in adults with tuberous sclerosis. While benign, these tumors are capable of provoking life threatening hemorrhage and replacement of the kidney parenchyma, resulting in renal failure. The histogenesis of these tumors is currently unclear, although currently, we believe these tumors arise from "perivascular epithelioid cells" of which no normal counterpart has been convincingly demonstrated. Recently, stem cell precursors have been recognized that can give rise to smooth muscle and melanocytes. These precursors have been shown to express the neural stem cell marker NG2 and L1. In order to determine whether angiomyolipomas, which exhibit smooth muscle and melanocytic phenotypes, express NG2 and L1, we performed immunocytochemistry on a cell line derived from a human angiomyolipoma, and found that these cells are uniformly positive. Immunohistochemistry of human angiomyolipoma specimens revealed uniform staining of tumor cells, while renal cell carcinomas revealed positivity only of angiogenic vessels. These results support a novel histogenesis of angiomyolipoma as a defect in differentiation of stem cell precursors.
Collapse
Affiliation(s)
- So Dug Lim
- Department of Urology, Emory University School of Medicine, and Atlanta VA Medical Center, Georgia 30322, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yamanaka H, Obata K, Kobayashi K, Dai Y, Fukuoka T, Noguchi K. Alteration of the cell adhesion molecule L1 expression in a specific subset of primary afferent neurons contributes to neuropathic pain. Eur J Neurosci 2007; 25:1097-111. [PMID: 17331206 PMCID: PMC1891330 DOI: 10.1111/j.1460-9568.2007.05344.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The cell adhesion molecule L1 (L1-CAM) plays important functional roles in the developing and adult nervous systems. Here we show that peripheral nerve injury induced dynamic post-transcriptional alteration of L1-CAM in the rat dorsal root ganglia (DRGs) and spinal cord. Sciatic nerve transection (SCNT) changed the expression of L1-CAM protein but not L1-CAM mRNA. In DRGs, SCNT induced accumulation of the L1-CAM into the surface of somata, which resulted in the formation of immunoreactive ring structures in a number of unmyelinated C-fiber neurons. These neurons with L1-CAM-immunoreactive ring structures were heavily colocalized with phosphorylated p38 MAPK. Western blot analysis revealed the increase of full-length L1-CAM and decrease of fragments of L1-CAM after SCNT in DRGs. Following SCNT, L1-CAM-immunoreactive profiles in the dorsal horn showed an increase mainly in pre-synaptic areas of laminae I–II with a delayed onset and colocalized with growth-associated protein 43. In contrast to DRGs, SCNT increased the proteolytic 80-kDa fragment of L1-CAM and decreased full-length L1-CAM in the spinal cord. The intrathecal injection of L1-CAM antibody for the extracellular domain of L1-CAM inhibited activation of p38 MAPK and emergence of ring structures of L1-CAM immunoreactivity in injured DRG neurons. Moreover, inhibition of extracellular L1-CAM binding by intrathecal administration of antibody suppressed the mechanical allodynia and thermal hyperalgesia induced by partial SCNT. Collectively, these data suggest that the modification of L1-CAM in nociceptive pathways might be an important pathomechanism of neuropathic pain.
Collapse
Affiliation(s)
- Hiroki Yamanaka
- Department of Anatomy and Neuroscience, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Mann F, Rougon G. Mechanisms of axon guidance: membrane dynamics and axonal transport in semaphorin signalling. J Neurochem 2007; 102:316-23. [PMID: 17442048 DOI: 10.1111/j.1471-4159.2007.04578.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The intricate geometry of neuronal networks poses many unique cell-biological problems regarding the way a growing axon responds to its environment. Several groups of ligand-receptor pairs have been identified to regulate such processes. In this study, we take class 3 semaphorins as an example and review what is known about the intracellular movements of semaphorins throughout neuronal cells, transport support structures and location of release sites. We discuss how their receptor trafficking may contribute to regulate membrane dynamics underlying growth cone motility and the physiological contribution made by class 3 semaphorins-induced acceleration of axoplasmic transport on neurite development.
Collapse
Affiliation(s)
- Fanny Mann
- Developmental Biology Institute of Marseille Luminy, CNRS UMR 6216, Université de la Méditerranée, Parc Scientifique de Luminy, Marseille cedex, France
| | | |
Collapse
|
41
|
Blackmore M, Letourneau PC. L1, beta1 integrin, and cadherins mediate axonal regeneration in the embryonic spinal cord. ACTA ACUST UNITED AC 2007; 66:1564-83. [PMID: 17058193 DOI: 10.1002/neu.20311] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Embryonic birds and mammals are capable of axon regeneration after spinal cord injury, but this ability is lost during a discrete developmental transition. We recently showed that changes within maturing neurons, as opposed to changes solely in the spinal cord environment, significantly restrict axon regeneration during development. The developmental changes within neurons that limit axon regeneration remain unclear. One gap in knowledge is the identity of the adhesive receptors that embryonic neurons use to extend axons in the spinal cord. Here we test the roles of L1/NgCAM, beta1 integrin, and cadherins, using a coculture system in which embryonic chick brainstem neurons regenerate axons into an explant of embryonic spinal cord. By in vivo and in vitro methods, we found that brainstem neurons reduce axonal expression of L1 as they mature. Disrupting either L1 or beta1 integrin function individually in our coculture system partially inhibited growth of brainstem axons in spinal cords, while disrupting cadherin function alone had no effect. However, when all three adhesive receptors were blocked simultaneously, axon growth in the spinal cord was reduced by 90%. Using immunohistochemistry and in situ hybridization we show that during the period when neurons lose their regenerative capacity they reduce expression of mRNA for N-cadherin, and reduce axonal L1/NgCAM protein through a post-transcriptional mechanism. These data show that embryonic neurons use L1/NgCAM, beta1 integrin, and cadherin receptors for axon regeneration in the embryonic spinal cord, and raise the possibility that a reduced expression of these essential receptors may contribute to the low-regenerative capacity of older neurons.
Collapse
Affiliation(s)
- Murray Blackmore
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | |
Collapse
|
42
|
Derycke L, Morbidelli L, Ziche M, De Wever O, Bracke M, Van Aken E. Soluble N-cadherin fragment promotes angiogenesis. Clin Exp Metastasis 2006; 23:187-201. [PMID: 17028923 DOI: 10.1007/s10585-006-9029-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Accepted: 06/21/2006] [Indexed: 10/24/2022]
Abstract
Endothelial cells express two dependent intercellular adhesion molecules: vascular endothelial (VE)-cadherin, specific for endothelial cells, and N-cadherin, also present in neuronal, lens, skeletal and heart muscle cells, osteoblasts, pericytes and fibroblasts. While there exists a vast amount of evidence that VE-cadherin promotes angiogenesis, the role of N-cadherin still remains to be elucidated. We found that a soluble 90-kDa fragment N-cadherin promotes angiogenesis in the rabbit cornea assay and in the chorioallantoic assay when cleaved enzymatically from the extracellular domain of N-cadherin. Soluble N-cadherin stimulates migration of endothelial cells in the wound healing assay and stimulates phosphorylation of extracellular regulated kinase. In vitro experiments with PD173074 and knock-down of N-cadherin and fibroblast growth factor (FGF)-receptor, showed that the pro-angiogenic effect of soluble N-cadherin is N-cadherin- and FGF-receptor-dependent. Our results suggest that soluble N-cadherin stimulates migration of endothelial cells through the FGF-receptor.
Collapse
Affiliation(s)
- L Derycke
- Laboratory of Experimental Cancerology, Department of Radiotherapy and Nuclear Medicine, Ghent University Hospital, De Pintelaan 185, 9000, Gent, Belgium.
| | | | | | | | | | | |
Collapse
|
43
|
Davies JE, Tang X, Bournat JC, Davies SJA. Decorin promotes plasminogen/plasmin expression within acute spinal cord injuries and by adult microglia in vitro. J Neurotrauma 2006; 23:397-408. [PMID: 16629625 DOI: 10.1089/neu.2006.23.397] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Spinal cord scar tissue presents a combined physical and molecular barrier to axon regeneration. Theoretically, spinal cord injuries (SCIs) can be rendered more permissive to axon growth by either suppressing synthesis of misaligned, fibrotic scar tissue and associated axon growth inhibitors, or enzymatically degrading them. We have previously shown that acute infusion of human recombinant decorin core protein into discreet stab injuries of the rat dorsal column pathways effected a major suppression of inflammation, astrogliosis, and multiple axon growth inhibitory chondroitin sulfate proteoglycans, which combined to promote rapid axon growth across the injury site. The high efficiency of chondroitin sulfate proteoglycan (CSPG) core protein suppression (approximately 90%) suggested that decorin may promote CSPG degradation in addition to suppressing CSPG synthesis. As the serine protease plasmin can degrade axon growth inhibitory CSPGs (neurocan and phosphacan) and its zymogen, plasmininogen is synthesized by microglia, we have investigated whether decorin treatment of acute SCIs and cultured adult spinal cord microglia can increase plasminogen/ plasmin synthesis. Infusion of hr-decorin over the first 8 days post-SCI induced 10- and 17-fold increases in plasminogen and plasmin protein levels, respectively, within sites of injury and a threefold increase in microglial plasminogen mRNA in vitro. In addition to potentially degrading multiple axon growth inhibitory components of the glial scar, plasmin is known to play major roles in activating neurotrophins and promoting central nervous system (CNS) plasticity. The wider implications of decorin induction of plasmin in the injured spinal cord for axon regeneration, and recovery of function at acute and chronic time points post-SCI are reviewed.
Collapse
Affiliation(s)
- Jeannette E Davies
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
44
|
Arlt MJE, Novak-Hofer I, Gast D, Gschwend V, Moldenhauer G, Grünberg J, Honer M, Schubiger PA, Altevogt P, Krüger A. Efficient inhibition of intra-peritoneal tumor growth and dissemination of human ovarian carcinoma cells in nude mice by anti-L1-cell adhesion molecule monoclonal antibody treatment. Cancer Res 2006; 66:936-43. [PMID: 16424028 DOI: 10.1158/0008-5472.can-05-1818] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The L1 cell adhesion molecule is implicated in the control of proliferation, migration, and invasion of several tumor cell types in vitro. Recently, L1 overexpression was found to correlate with tumor progression of ovarian carcinoma, one of the most common causes of cancer-related deaths in gynecologic malignant diseases. To evaluate L1 as a potential target for ovarian cancer therapy, we investigated the effects of anti-L1 monoclonal antibodies (chCE7 and L1-11A) on proliferation and migration of L1-positive human SKOV3ip ovarian carcinoma cells in vitro and the therapeutic efficacy of L1-11A against i.p. SKOV3ip tumor growth in nude mice. In vitro, both anti-L1 antibodies efficiently inhibited the proliferation of SKOV3ip cells as well as other L1-expressing tumor cell lines (renal carcinoma, neuroblastoma, and colon carcinoma). On two cell lines, hyper-cross-linking of L1-11A with a secondary antibody was necessary for significant inhibition of proliferation, indicating that cross-linking of L1 is required for the antiproliferative effect. L1-negative prostate carcinoma cells were not influenced by antibody treatment. Biweekly treatment of ovarian carcinoma-bearing mice with L1-11A led to a dose-dependent and significant reduction of tumor burden (up to -63.5%) and ascites formation (up to -75%). This effect was associated with reduced proliferation within the tumors. L1-directed antibody-based inhibition of peritoneal growth and dissemination of human ovarian carcinoma cells represents important proof-of-principle for the development of a new therapy against one of the leading gynecologic malignant diseases.
Collapse
Affiliation(s)
- Matthias J E Arlt
- Klinikum rechts der Isar der Technischen Universität München, Institut für Experimentelle Onkologie und Therapieforschung, Ismaninger Strasse 22, D-81675 Munich, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Strekalova H, Buhmann C, Kleene R, Eggers C, Saffell J, Hemperly J, Weiller C, Müller-Thomsen T, Schachner M. Elevated levels of neural recognition molecule L1 in the cerebrospinal fluid of patients with Alzheimer disease and other dementia syndromes. Neurobiol Aging 2006; 27:1-9. [PMID: 16298234 DOI: 10.1016/j.neurobiolaging.2004.11.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2004] [Revised: 10/26/2004] [Accepted: 11/18/2004] [Indexed: 11/20/2022]
Abstract
In this study we surveyed a total of 218 cerebrospinal fluid (CSF) samples from patients with different neurological diseases including Alzheimer disease, non-Alzheimer forms of dementia, other neurodegenerative diseases without dementia and normal controls to quantitate by capture ELISA the concentrations of the immunoglobulin superfamily adhesion molecules L1 and NCAM, and characterized by immunoblot analysis the molecular forms of L1 and NCAM. We found a significant increase of L1 and a strong tendency for increase of the soluble fragments of NCAM in the CSF of Alzheimer patients compared to the normal control group. The proteolytic fragments of L1, but not NCAM were also elevated in patients with vascular dementia and dementia of mixed type. Higher L1 concentrations were observed irrespective of age and gender. NCAM concentrations were independent of gender, but positively correlated with age and, surprisingly, also with incidence of multiple sclerosis. Thus, there was an influence of Alzheimer and non-Alzheimer dementias and neurodegeneration on L1, whereas age and neurodegeneration influenced NCAM concentrations. These observations point to an abnormal processing and/or shedding of L1 and NCAM in dementia-related neurodegeneration and age, respectively, reflecting changes in adhesion molecule-related cell interactions.
Collapse
Affiliation(s)
- Helen Strekalova
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Martinistr. 52, D 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hall H, Djonov V, Ehrbar M, Hoechli M, Hubbell JA. Heterophilic interactions between cell adhesion molecule L1 and alphavbeta3-integrin induce HUVEC process extension in vitro and angiogenesis in vivo. Angiogenesis 2005; 7:213-23. [PMID: 15609076 DOI: 10.1007/s10456-004-1328-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 07/07/2004] [Indexed: 02/06/2023]
Abstract
Cell adhesion molecule L1 was implicated in angiogenic processes, tumor formation and metastasis. Here, we provide evidence that the sixth Ig-like domain of L1 (L1Ig6) interacts with alpha(v)beta3 to induce process extension of human umbilical vein endothelial cells (HUVECs) in vitro and angiogenesis in vivo. HUVECs formed network-like structures on full-length L1 or L1Ig6 substrates comparable to structures found on matrigel. In the presence of mab alpha(v)beta3 or cyclic RGD, apoptosis was induced. In fibrin matrices where L1Ig6 was covalently incorporated, HUVECs formed multicellular and hollow processes through interactions between cell-surface alpha(v)beta3 and RGD-sites of matrix-immobilized L1Ig6. No such processes were induced by L1Ig6 having non-functional RDG-sites, or in the presence of mab alpha(v)beta3 or cyclic RGD. In those matrices, increased apoptosis was found. Co-immunoprecipitation of L1 or L1Ig6 with alpha(v)beta3 suggests close interactions. Furthermore, L1Ig6 stimulated HUVECs showed increased tyrosine phosphorylation of alpha(v)beta3 and phosphorylation of MAP kinases (ERK1 and ERK2) but not AKT indicating specific activation of alpha(v) and alpha(v)beta3 followed by activation of downstream kinases. Application of L1Ig6-modified fibrin matrices on CAMs induced 50-60% increased alpha(v) and alpha(v)beta3 protein expression and in vivo angiogenesis indicated by approximately 50% increased mean vascular length density. The results demonstrate angiogenic potential of L1Ig6 involving ligation and activation of alpha(v)beta3.
Collapse
Affiliation(s)
- Heike Hall
- Institute for Biomedical Engineering and Department of Materials, ETH and University of Zurich, Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
47
|
Loers G, Chen S, Grumet M, Schachner M. Signal transduction pathways implicated in neural recognition molecule L1 triggered neuroprotection and neuritogenesis. J Neurochem 2005; 92:1463-76. [PMID: 15748164 DOI: 10.1111/j.1471-4159.2004.02983.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The signal transduction pathways involved in adhesion molecule L1-triggered neuritogenesis and neuroprotection were investigated using the extracellular domain of mouse or human L1 in fusion with the Fc portion of human immunoglobulin G or L1 purified from mouse brain by affinity chromatography. Substrate L1-triggered neuritogenesis and neuroprotection depended on distinct but also overlapping signal transduction pathways and on the expression of L1 at the neuronal cell surface. PI3 kinase inhibitors, Src family kinase inhibitors as well as mitogen-activated protein kinase kinase inhibitors reduced both L1-triggered neuritogenesis and neuroprotection. In contrast, fibroblast growth factor receptor inhibitors, a protein kinase A inhibitor, and an inhibitor of cAMP-mediated signal transduction pathways, blocked neuritogenesis, but did not affect L1-triggered neuroprotection. Proteolytic cleavage of L1 or its interaction partners is necessary for both L1-mediated neuritogensis and neuroprotection. Furthermore, L1-triggered neuroprotection was found to be associated with increased phosphorylation of extracellular signal-regulated kinases 1/2, Akt and Bad, and inhibition of caspases. These observations suggest possibilities of differentially targeting signal transduction pathways for L1-dependent neuritogenesis and neuroprotection.
Collapse
Affiliation(s)
- Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Falkenried 94, D-20251 Hamburg, Germany
| | | | | | | |
Collapse
|
48
|
Allory Y, Matsuoka Y, Bazille C, Christensen EI, Ronco P, Debiec H. The L1 Cell Adhesion Molecule Is Induced in Renal Cancer Cells and Correlates with Metastasis in Clear Cell Carcinomas. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.1190.11.3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: The L1 cell adhesion molecule is overexpressed in many human carcinomas. The objectives of the study were to provide a comprehensive description of L1 distribution in human kidney and to establish the prognostic relevance of L1 expression in renal cell carcinomas (RCC).
Experimental Design: Using two antibodies to the extracellular part and the cytoplasmic domain, respectively, we first compared L1 expression in normal kidney and renal tumors of diverse histopathologic origin, then we studied L1 expression together with tumor stage, grade, molecular prognostic biomarkers, and metastatic behavior.
Results: In normal kidney, L1 immunoreactive with both antibodies was expressed in all epithelial cells originating from the ureteric bud except for intercalated cells. In renal tumors, L1 was mainly detected in those originating from cells that do not express L1 in the normal kidney [i.e., 33 of 72 clear cell RCC (ccRCC) and 25 of 88 papillary RCC (papRCC)]. Both in ccRCC and papRCC, L1 reacted only with the antibody to the extracellular domain, suggesting that the protein was truncated. In these carcinomas, L1 expression was strongly correlated with Ki-67 proliferation index (ccRCC, P = 0.0059; papRCC, P = 0.0039), but only in ccRCC, the presence of L1 was associated with the risk of metastasis (P = 0.0121). This risk was higher if cyclin D1 was concurrently absent in tumor cells (P < 0.0001). The L1+/cyclin D1− profile was an independent prognostic factor of metastasis occurrence in multivariate analysis (P = 0.0023).
Conclusion: We have found a combination of markers that can serve to identify a subgroup of high-risk patients with ccRCC that may require more aggressive therapies.
Collapse
Affiliation(s)
- Yves Allory
- 1Institut National de la Santé et de la Recherche Médicale U489 and
| | - Yasuko Matsuoka
- 1Institut National de la Santé et de la Recherche Médicale U489 and
| | - Céline Bazille
- 2Department of Pathology, Tenon Hospital (Assistance Publique-Hôpitaux de Paris) and Paris 6 University, Paris, France and
| | | | - Pierre Ronco
- 1Institut National de la Santé et de la Recherche Médicale U489 and
| | - Hanna Debiec
- 1Institut National de la Santé et de la Recherche Médicale U489 and
| |
Collapse
|
49
|
Castellani V, Falk J, Rougon G. Semaphorin3A-induced receptor endocytosis during axon guidance responses is mediated by L1 CAM. Mol Cell Neurosci 2004; 26:89-100. [PMID: 15121181 DOI: 10.1016/j.mcn.2004.01.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 01/09/2004] [Accepted: 01/16/2004] [Indexed: 11/22/2022] Open
Abstract
During axon navigation, Semaphorin3A-induced growth cone retraction is correlated with endocytosis. Although its function remains elusive, we showed previously that the cell adhesion molecule of the immunoglobulin super family L1 associates with Neuropilin-1 (NP-1) the Sema3A-binding subunit of the receptor complex and is required for Sema3A to elicit axonal repulsive responses. We report here that upon Sema3A binding to NP-1, L1 and NP-1 are co-internalized through a clathrin-dependent mechanism mediated by L1. We show that in COS7 cells, L1/NP-1 endocytosis is correlated with a cell contraction similar to that observed with the Plexin (Plex)/NP-1 or Plex/NP1/L1 complexes. In neuronal cultures, a L1-mimetic peptide able to switch Sema3A repulsive responses to attraction blocks both endocytosis and growth cone collapse. Similarly, in the COS7 cell model, peptide application prevents both the Sema3-induced L1/NP-1 internalization and cell collapse. These studies demonstrate that the L1/NP-1 complex is able to confer a biological response to Sema3A with L1 mediating receptor internalization following ligand activation. They also reveal that endocytosis controlled by L1/NP-1 cis and trans interactions is pivotal in Sema3A-mediated axon guidance.
Collapse
Affiliation(s)
- Valérie Castellani
- Laboratoire de Neurogenèse et Morphogenèse dans le Développement et chez l'Adulte; UMR CNRS 6156, Université de la Méditerranée, IBDM, 13288 Marseilles 9, France
| | | | | |
Collapse
|
50
|
Frappé I, Wang C, Caines G, Rideout-Gros S, Aubert I. Cell adhesion molecule L1 promotes neurite outgrowth of septal neurons. J Neurosci Res 2004; 75:667-77. [PMID: 14991842 DOI: 10.1002/jnr.20026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To establish if the cell adhesion molecule L1 could promote neurite outgrowth of septal neurons, L1-positive substrates were prepared by genetically modifying 3T3 fibroblasts with a retroviral vector encoding human L1 under the control of a negative tetracycline-regulatory system. In several clones of L1-transfected fibroblasts, L1 expression at the cell surface was prominent and efficiently regulated by doxycycline, a tetracycline analogue. In co-culture of septal neurons and fibroblasts, a two-dimensional fractionator probe provided systematic random sampling of the neurites to be measured. Septal neurons, isolated at embryonic Day 17, were found to express L1 in vitro and to extend significantly longer neurites when plated on L1-expressing fibroblasts compared to control fibroblasts. The neurite outgrowth-promoting effect of L1 was inhibited after a doxycycline treatment, which specifically suppressed L1 expression from the modified fibroblasts. The findings that septal neurons at embryonic Day 17 in vitro express L1 and respond to L1-modulation suggest that this molecule is involved in development of the septohippocampal pathway.
Collapse
Affiliation(s)
- Isabelle Frappé
- Neuroscience Research, Sunnybrook and Women's College Health Sciences Centre, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|