Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
1.
Sci Rep ; 14(1): 13560, 2024 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866905

RESUMEN

L1CAM-positive extracellular vesicles (L1EV) are an emerging biomarker that may better reflect ongoing neuronal damage than other blood-based biomarkers. The physiological roles and regulation of L1EVs and their small RNA cargoes following stroke is unknown. We sought to characterize L1EV small RNAs following stroke and assess L1EV RNA signatures for diagnosing stroke using weighted gene co-expression network analysis and random forest (RF) machine learning algorithms. Interestingly, small RNA sequencing of plasma L1EVs from patients with stroke and control patients (n = 28) identified micro(mi)RNAs known to be enriched in the brain. Weighted gene co-expression network analysis (WGCNA) revealed small RNA transcript modules correlated to diagnosis, initial NIH stroke scale, and age. L1EV RNA signatures associated with the diagnosis of AIS were derived from WGCNA and RF classification. These small RNA signatures demonstrated a high degree of accuracy in the diagnosis of AIS with an area under the curve (AUC) of the signatures ranging from 0.833 to 0.932. Further work is necessary to understand the role of small RNA L1EV cargoes in the response to brain injury, however, this study supports the utility of L1EV small RNA signatures as a biomarker of stroke.


Asunto(s)
Biomarcadores , Vesículas Extracelulares , Accidente Cerebrovascular Isquémico , Molécula L1 de Adhesión de Célula Nerviosa , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , Masculino , Accidente Cerebrovascular Isquémico/genética , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/diagnóstico , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Femenino , Anciano , Biomarcadores/sangre , Persona de Mediana Edad , Aprendizaje Automático , MicroARNs/genética , MicroARNs/sangre , MicroARNs/metabolismo
2.
Cell Biochem Funct ; 42(4): e4034, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38715189

RESUMEN

L1 syndrome, a neurological disorder with an X-linked inheritance pattern, mainly results from mutations occurring in the L1 cell adhesion molecule (L1CAM) gene. The L1CAM molecule, belonging to the immunoglobulin (Ig) superfamily of neurocyte adhesion molecules, plays a pivotal role in facilitating intercellular signal transmission across membranes and is indispensable for proper neuronal development and function. This study identified a rare missense variant (c.1759G>C; p.G587R) in the L1CAM gene within a male fetus presenting with hydrocephalus. Due to a lack of functional analysis, the significance of the L1CAM mutation c.1759G>C (p.G587R) remains unknown. We aimed to perform further verification for its pathogenicity. Blood samples were obtained from the proband and his parents for trio clinical exome sequencing and mutation analysis. Expression level analysis was conducted using western blot techniques. Immunofluorescence was employed to investigate L1CAM subcellular localization, while cell aggregation and cell scratch assays were utilized to assess protein function. The study showed that the mutation (c.1759G>C; p.G587R) affected posttranslational glycosylation modification and induced alterations in the subcellular localization of L1-G587R in the cells. It resulted in the diminished expression of L1CAM on the cell surface and accumulation in the endoplasmic reticulum. The p.G587R altered the function of L1CAM protein and reduced homophilic adhesion capacity of proteins, leading to impaired adhesion and migration of proteins between cells. Our findings provide first biological evidence for the association between the missense mutation (c.1759G>c; p.G587R) in the L1CAM gene and L1 syndrome, confirming the pathogenicity of this missense mutation.


Asunto(s)
Mutación Missense , Molécula L1 de Adhesión de Célula Nerviosa , Humanos , Masculino , Células HEK293 , Hidrocefalia/genética , Hidrocefalia/metabolismo , Hidrocefalia/patología , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Linaje , Recién Nacido
3.
Biomolecules ; 14(4)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38672483

RESUMEN

The X-chromosome-linked cell adhesion molecule L1 (L1CAM), a glycoprotein mainly expressed by neurons in the central and peripheral nervous systems, has been implicated in many neural processes, including neuronal migration and survival, neuritogenesis, synapse formation, synaptic plasticity and regeneration. L1 consists of extracellular, transmembrane and cytoplasmic domains. Proteolytic cleavage of L1's extracellular and transmembrane domains by different proteases generates several L1 fragments with different functions. We found that myelin basic protein (MBP) cleaves L1's extracellular domain, leading to enhanced neuritogenesis and neuronal survival in vitro. To investigate in vivo the importance of the MBP-generated 70 kDa fragment (L1-70), we generated mice with an arginine to alanine substitution at position 687 (L1/687), thereby disrupting L1's MBP cleavage site and obliterating L1-70. Young adult L1/687 males showed normal anxiety and circadian rhythm activities but enhanced locomotion, while females showed altered social interactions. Older L1/687 males were impaired in motor coordination. Furthermore, L1/687 male and female mice had a larger hippocampus, with more neurons in the dentate gyrus and more proliferating cells in the subgranular layer, while the thickness of the corpus callosum and the size of lateral ventricles were normal. In summary, subtle mutant morphological changes result in subtle behavioral changes.


Asunto(s)
Encéfalo , Molécula L1 de Adhesión de Célula Nerviosa , Animales , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Ratones , Masculino , Femenino , Encéfalo/metabolismo , Fibronectinas/metabolismo , Fibronectinas/genética , Mutación , Conducta Animal , Dominios Proteicos , Neuronas/metabolismo , Hipocampo/metabolismo , Ratones Endogámicos C57BL
4.
Brain Dev ; 46(6): 230-233, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38480026

RESUMEN

BACKGROUND: Heterozygous L1CAM variants cause L1 syndrome with hydrocephalus and aplasia/hypoplasia of the corpus callosum. L1 syndrome usually has an X-linked recessive inheritance pattern; however, we report a rare case occurring in a female child. CASE PRESENTATION: The patient's family history was unremarkable. Fetal ultrasonography revealed enlarged bilateral ventricles of the brain and hypoplasia of the corpus callosum. The patient was born at 38 weeks and 4 days of gestation. Brain MRI performed on the 8th day of life revealed enlargement of the brain ventricles, marked in the lateral and third ventricles with irregular margins, and hypoplasia of the corpus callosum. Exome sequencing at the age of 2 years and 3 months revealed a de novo heterozygous L1CAM variant (NM_000425.5: c.2934_2935delp. (His978Glnfs * 25). X-chromosome inactivation using the human androgen receptor assay revealed that the pattern of X-chromosome inactivation in the patients was highly skewed (96.6 %). The patient is now 4 years and 11 months old and has a mild developmental delay (developmental quotient, 56) without significant progression of hydrocephalus. CONCLUSION: In this case, we hypothesized that the dominant expression of the variant allele arising from skewed X inactivation likely caused L1 syndrome. Symptomatic female carriers may challenge the current policies of prenatal and preimplantation diagnoses.


Asunto(s)
Hidrocefalia , Molécula L1 de Adhesión de Célula Nerviosa , Inactivación del Cromosoma X , Humanos , Femenino , Inactivación del Cromosoma X/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Hidrocefalia/genética , Hidrocefalia/diagnóstico por imagen , Preescolar , Agenesia del Cuerpo Calloso/genética
5.
Am J Clin Pathol ; 162(1): 75-85, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38430556

RESUMEN

OBJECTIVES: We sought to confirm utility of our institution's modified Proactive Molecular Risk Classifier for Endometrial Cancer protocol in our daily practice, which includes mismatch repair (MMR), p53, and L1 cell adhesion molecule (L1CAM) immunohistochemistry with in-house next-generation sequencing for POLE, TP53, and CTNNB1. METHODS: We conducted a retrospective review of all patients in our institution who underwent primary endometrial carcinoma resection from the year prior to protocol implementation (PRE; October 1, 2020, to September 30, 2021) through first year of implementation (POST; October 1, 2021, to September 30, 2022) to compare the distribution of molecular and traditional staging factors using GOG-249 criteria to assign clinical risk. RESULTS: In total, 136 of 260 PRE patients were classified as clinically low risk (LR), of whom 31 were MMR deficient. Of the 157 LR POST patients with endometrioid-type carcinoma, 45 were MMR deficient, 5 were POLE mutant, 5 were TP53 mutant, 56 were of no specific molecular profile (NSMP), and 46 did not receive full protocol testing. Of all 79 POST NSMP endometrioid-type cases, 18 were CTNNB1 mutated and 8 showed L1CAM expression. CONCLUSIONS: Our protocol identified 22 (14%) of 157 LR tumors that harbored incipient intermediate- to high-risk molecular aberrations in TP53, CTNNB1, or L1CAM. Moving forward, results of ongoing trials assessing adjuvant therapy decisions based on molecular classification are necessary to confirm protocol utility and identify appropriate modifications.


Asunto(s)
Neoplasias Endometriales , Proteína p53 Supresora de Tumor , Humanos , Femenino , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Estudios Retrospectivos , Persona de Mediana Edad , Anciano , Proteína p53 Supresora de Tumor/genética , Biomarcadores de Tumor/genética , Reparación de la Incompatibilidad de ADN/genética , beta Catenina/genética , Inmunohistoquímica , Adulto , Secuenciación de Nucleótidos de Alto Rendimiento , Anciano de 80 o más Años , Molécula L1 de Adhesión de Célula Nerviosa/genética , Carcinoma Endometrioide/genética , Carcinoma Endometrioide/patología
6.
Mod Pathol ; 37(5): 100467, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38460672

RESUMEN

Renal low-grade oncocytic tumor (LOT) is a recently recognized renal cell neoplasm designated within the "other oncocytic tumors" category in the 2022 World Health Organization classification system. Although the clinicopathologic, immunohistochemical, and molecular features reported for LOT have been largely consistent, the data are relatively limited. The morphologic overlap between LOT and other low-grade oncocytic neoplasms, particularly eosinophilic chromophobe renal cell carcinoma (E-chRCC), remains a controversial area in renal tumor classification. To address this uncertainty, we characterized and compared large cohorts of LOT (n = 67) and E-chRCC (n = 69) and revealed notable differences between the 2 entities. Clinically, LOT predominantly affected women, whereas E-chRCC showed a male predilection. Histologically, although almost all LOTs were dominated by a small-nested pattern, E-chRCC mainly showed solid and tubular architectures. Molecular analysis revealed that 87% of LOT cases harbored mutations in the tuberous sclerosis complex (TSC)-mTOR complex 1 (mTORC1) pathway, most frequently in MTOR and RHEB genes; a subset of LOT cases had chromosomal 7 and 19q gains. In contrast, E-chRCC lacked mTORC1 mutations, and 60% of cases displayed chromosomal losses characteristic of chRCC. We also explored the cell of origin for LOT and identified L1 cell adhesion molecule (L1CAM), a collecting duct and connecting tubule principal cell marker, as a highly sensitive and specific ancillary test for differentiating LOT from E-chRCC. This distinctive L1CAM immunohistochemical labeling suggests the principal cells as the cell of origin for LOT, unlike the intercalated cell origin of E-chRCC and oncocytoma. The ultrastructural analysis of LOT showed normal-appearing mitochondria and intracytoplasmic lumina with microvilli, different from what has been described for chRCC. Our study further supports LOT as a unique entity with a benign clinical course. Based on the likely cell of origin and its clinicopathologic characteristics, we propose that changing the nomenclature of LOT to "Oncocytic Principal Cell Adenoma of the Kidney" may be a better way to define and describe this entity.


Asunto(s)
Adenoma Oxifílico , Biomarcadores de Tumor , Carcinoma de Células Renales , Neoplasias Renales , Molécula L1 de Adhesión de Célula Nerviosa , Humanos , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/química , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/química , Femenino , Masculino , Persona de Mediana Edad , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/análisis , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Anciano , Adulto , Adenoma Oxifílico/patología , Adenoma Oxifílico/genética , Diagnóstico Diferencial , Anciano de 80 o más Años , Inmunohistoquímica , Clasificación del Tumor , Mutación
7.
Int J Mol Sci ; 25(5)2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38474289

RESUMEN

The L1 cell adhesion molecule (L1) has demonstrated a range of beneficial effects in animal models of spinal cord injury, neurodegenerative disease, and ischemia; however, the role of L1 in TBI has not been fully examined. Mutations in the L1 gene affecting the extracellular domain of this type 1 transmembrane glycoprotein have been identified in patients with L1 syndrome. These patients suffer from hydrocephalus, MASA (mental retardation, adducted thumbs, shuffling gait, aphasia) symptoms, and corpus callosum agenesis. Clinicians have observed that recovery post-traumatic brain injury (TBI) varies among the population. This variability may be explained by the genetic differences present in the general population. In this study, we utilized a novel mouse model of L1 syndrome with a mutation at aspartic acid position 201 in the extracellular domain of L1 (L1-201). We assessed the impact of this specific single nucleotide polymorphism (SNP) localized to the X-chromosome L1 gene on recovery outcomes following TBI by comparing the L1-201 mouse mutants with their wild-type littermates. We demonstrate that male L1-201 mice exhibit significantly worse learning and memory outcomes in the Morris water maze after lateral fluid percussion (LFP) injury compared to male wild-type mice and a trend to worse motor function on the rotarod. However, no significant changes were observed in markers for inflammatory responses or apoptosis after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Enfermedades Genéticas Ligadas al Cromosoma X , Hidrocefalia , Discapacidad Intelectual , Molécula L1 de Adhesión de Célula Nerviosa , Enfermedades Neurodegenerativas , Paraplejía Espástica Hereditaria , Humanos , Masculino , Animales , Ratones , Molécula L1 de Adhesión de Célula Nerviosa/genética , Polimorfismo de Nucleótido Simple , Hidrocefalia/genética
8.
Gynecol Oncol ; 184: 132-138, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38309030

RESUMEN

OBJECTIVE: We investigate the prognostic role of ß-catenin and L1 neuronal cell-adhesion molecule (L1CAM) according to risk groups in endometrial carcinomas (EC). METHODS: A total of 335 EC patients were classified according to the Proactive Molecular Risk Classifier for Endometrial Cancer. We evaluated the expression of ß-catenin and L1CAM using immunohistochemistry, and their association with clinicopathological characteristics and survival. RESULTS: The expressions of ß-catenin and L1CAM were observed in 10.4% of all patients, respectively, and showed mutually exclusive pattern. While ß-catenin expression was associated with endometrioid histology (p = 0.035) and low tumor grade (p = 0.045), L1CAM expression was associated with non-endometrioid histology (p < 0.001), high tumor grade (p < 0.001), lymphovascular space invasion (p = 0.006), and advanced International Federation of Gynecology and Obstetrics (FIGO) stage (p = 0.001). ß-catenin expression was most frequent in the no specific molecular (NSMP) group (26/35, 74.3%), followed by the DNA polymerase-ε-mutated (POLE-mut) (6/35, 17.1%), and mismatch repair-deficiency (dMMR) (3/35, 8.6%). L1CAM expression was most frequent in the p53-abnormal group (22/35, 62.9%), followed by the NSMP (6/35, 17.1%), dMMR (4/35, 11.4%), and POLE-mut (3/35, 8.6%). Although both markers did not show statistical significance in multivariate analysis for both progression-free survival (PFS) and overall survival in entire cohort, ß-catenin positivity was identified as the sole factor associated with worse PFS in the high-intermediate risk subgroup (p = 0.001). CONCLUSION: The expression of nuclear ß-catenin may serve as a potential biomarker for predicting recurrence and guiding therapeutic strategies in high-intermediate risk EC patients.


Asunto(s)
Neoplasias Endometriales , Molécula L1 de Adhesión de Célula Nerviosa , beta Catenina , Humanos , Femenino , Neoplasias Endometriales/patología , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Molécula L1 de Adhesión de Célula Nerviosa/genética , beta Catenina/metabolismo , beta Catenina/biosíntesis , beta Catenina/genética , Persona de Mediana Edad , Anciano , Pronóstico , Adulto , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Anciano de 80 o más Años , Carcinoma Endometrioide/patología , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/genética , Clasificación del Tumor , Estadificación de Neoplasias
9.
J Cell Sci ; 137(3)2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38206094

RESUMEN

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.


Asunto(s)
Calicreínas , Molécula L1 de Adhesión de Célula Nerviosa , Animales , Ratones , Hipocampo/metabolismo , Calicreínas/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Plasticidad Neuronal/fisiología , Serina Proteasas/metabolismo
10.
Breast Cancer Res Treat ; 204(3): 465-474, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38183514

RESUMEN

PURPOSE: The potential of targeting forkhead box C1 (FOXC1) as a therapeutic approach for triple-negative breast cancer (TNBC) is promising. However, a comprehensive understanding of FOXC1 regulation, particularly upstream factors, remains elusive. Expression of the L1 cell adhesion molecule (L1CAM), a transmembrane glycoprotein associated with brain metastasis, was observed to be positively associated with FOXC1 transcripts. Thus, this study aims to investigate their relationship in TNBC progression. METHODS: Publicly available FOXC1 and L1CAM transcriptomic data were obtained, and their corresponding proteins were analyzed in four TNBC cell lines. In BT549 cells, FOXC1 and L1CAM were individually silenced, while L1CAM was overexpressed in BT549-shFOXC1, MDA-MB-231, and HCC1937 cells. CCK-8, transwell, and wound healing assays were performed in these cell lines, and immunohistochemical staining was conducted in tumor samples. RESULTS: A positive correlation between L1CAM and FOXC1 transcripts was observed in publicly available datasets. In BT549 cells, knockdown of FOXC1 led to reduced L1CAM expression at both the transcriptional and protein levels, and conversely, silencing of L1CAM decreased FOXC1 protein levels, but interestingly, FOXC1 transcripts remained largely unaffected. Overexpressing L1CAM resulted in increased FOXC1 protein expression without significant changes in FOXC1 mRNA levels. This trend was also observed in BT549-shFOXC1, MDA-MB-231-L1CAM, and HCC1937-L1CAM cells. Notably, alterations in FOXC1 or L1CAM levels corresponded to changes in cell proliferation, migration, and invasion capacities. Furthermore, a positive correlation between L1CAM and FOXC1 protein expression was detected in human TNBC tumors. CONCLUSION: FOXC1 and L1CAM exhibit co-regulation at the protein level, with FOXC1 regulating at the transcriptional level and L1CAM regulating at the post-transcriptional level, and together they positively influence cell proliferation, migration, and invasion in TNBC.


Asunto(s)
Factores de Transcripción Forkhead , Molécula L1 de Adhesión de Célula Nerviosa , Neoplasias de la Mama Triple Negativas , Humanos , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/uso terapéutico , Neoplasias de la Mama Triple Negativas/patología
12.
Arch Gynecol Obstet ; 309(3): 789-799, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37454351

RESUMEN

INTRODUCTION: Molecular and genomic profiling in endometrial cancer is increasing popularity. L1 cell adhesion molecule (L1CAM) is frequently mutated in endometrial cancer. In this paper, we aim to evaluate the prognostic role of L1CAM in patients with stage I endometrial cancer. METHODS: We performed a systematic review and meta-analysis searching in PubMed (MEDLINE), EMBASE, and Web of Science database to identify studies reporting the expression of L1CAM in endometrial cancer. The primary endpoint measure was to assess and evaluate the impact of L1CAM on survival outcomes. This study was performed according to the Preferred Reporting Items for Systematic review and Meta-Analysis Protocols (PRISMA-P) statement. RESULTS: Five studies were included. The pooled results suggested that L1CAM expression influences survival outcomes in stage I endometrial cancer. High L1CAM expression correlated with worse disease-free survival (HR 4.11, 95% CI 1.02-16.59, p = 0.047) and overall survival (HR 3.62, 95% CI 1.32-9.31, p = 0.012). High L1CAM level was also associated with a more aggressive FIGO grade and with older age. CONCLUSION: This systematic review supported that L1CAM have a prognostic role in stage I endometrial cancer, thus providing a potential useful tool for tailoring the need of adjuvant therapy.


Asunto(s)
Neoplasias Endometriales , Molécula L1 de Adhesión de Célula Nerviosa , Femenino , Humanos , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Estadificación de Neoplasias , Biomarcadores de Tumor/genética , Revisiones Sistemáticas como Asunto , Metaanálisis como Asunto , Neoplasias Endometriales/patología , Pronóstico
13.
Int J Mol Sci ; 24(24)2023 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-38139155

RESUMEN

The vesicle-associated membrane protein 7 (VAMP7) is a SNARE protein of the longin family involved in a wide range of subcellular trafficking events, including neurite sprouting and elongation. The expression of the human gene SYBL1, encoding VAMP7, is finely regulated by alternative splicing. Among the minor isoforms identified so far, VAMP7j is the one most expressed and modulated in the human brain. Therefore, we focused on gaining functional evidence on VAMP7j, which lacks a functional SNARE motif but retains both the longin and transmembrane domains. In human SH-SY5Y cells, we found VAMP7j to modulate neuritogenesis by mediating transport of L1CAM toward the plasma membrane, in a fashion regulated by phosphorylation of the longin domain. VAMP7-mediated regulation of L1CAM trafficking seems at least to differentiate humans from rats, with VAMP7j CNS expression being restricted to primates, including humans. Since L1CAM is a central player in neuritogenesis and axon guidance, these findings suggest the species-specific splicing of SYBL1 is among the fine tuners of human neurodevelopmental complexity.


Asunto(s)
Molécula L1 de Adhesión de Célula Nerviosa , Neuroblastoma , Animales , Humanos , Ratas , Membrana Celular/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuroblastoma/metabolismo , Proyección Neuronal , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo
14.
PLoS One ; 18(11): e0294146, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37943774

RESUMEN

CRISPR/Cas9 is a powerful genome editing system that has remarkably facilitated gene knockout and targeted knock-in. To accelerate the practical use of CRISPR/Cas9, however, it remains crucial to improve the efficiency, precision, and specificity of genome editing, particularly targeted knock-in, achieved with this system. To improve genome editing efficiency, researchers should first have a molecular assay that allows sensitive monitoring of genome editing events with simple procedures. In the current study, we demonstrate that genome editing events occurring in L1CAM, an X-chromosome gene encoding a cell surface protein, can be readily monitored using flow cytometry (FCM) in multiple human cell lines including neuroblastoma cell lines. The abrogation of L1CAM was efficiently achieved using Cas9 nucleases which disrupt exons encoding the L1CAM extracellular domain, and was easily detected by FCM using anti-L1CAM antibodies. Notably, L1CAM-abrogated cells could be quantified by FCM in four days after transfection with a Cas9 nuclease, which is much faster than an established assay based on the PIGA gene. In addition, the L1CAM-based assay allowed us to measure the efficiency of targeted knock-in (correction of L1CAM mutations) accomplished through different strategies, including a Cas9 nuclease-mediated method, tandem paired nicking, and prime editing. Our L1CAM-based assay using FCM enables rapid and sensitive quantification of genome editing efficiencies and will thereby help researchers improve genome editing technologies.


Asunto(s)
Edición Génica , Molécula L1 de Adhesión de Célula Nerviosa , Humanos , Edición Génica/métodos , Citometría de Flujo , Sistemas CRISPR-Cas/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Línea Celular
15.
Anticancer Res ; 43(10): 4559-4571, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37772557

RESUMEN

BACKGROUND/AIM: The expression of L1 cell adhesion molecule (L1CAM) in uterine mesonephric-like adenocarcinoma (MLA) remains understudied. Our aim was to explore the L1CAM expression in uterine MLA, delving into its clinicopathological implications and prognostic significance. PATIENTS AND METHODS: We conducted L1CAM immunostaining in MLA, endometrioid carcinoma (EC), and serous carcinoma (SC), compared L1CAM expression across these histological types, and probed the relationship between L1CAM expression and the clinicopathological features and outcomes of patients with MLA. RESULTS: High L1CAM expression was evident in 15 of 28 MLA cases (53.6%). This rate surpassed that of EC (7.5%) but was less than that of SC (78.9%). A high L1CAM expression correlated with initial distant metastasis, advanced initial stage, lung metastasis, and the recurrence of MLA. L1CAM-high MLA exhibited worse disease-free and overall survival than L1CAM-low MLA. CONCLUSION: L1CAM over-expression was observed in more than half of the MLA cases, and was associated with aggressive clinicopathological traits and adverse outcomes in patients with uterine MLA.


Asunto(s)
Adenocarcinoma , Carcinoma Endometrioide , Neoplasias Endometriales , Molécula L1 de Adhesión de Célula Nerviosa , Femenino , Humanos , Pronóstico , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neoplasias Endometriales/patología , Biomarcadores de Tumor/metabolismo , Carcinoma Endometrioide/patología
16.
In Vivo ; 37(5): 2334-2339, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37652517

RESUMEN

AIM: Our study aimed to assess expression of L1 cell adhesion molecule (L1CAM) in early-stage cervical squamous-cell cancer as a prognostic factor. PATIENTS AND METHODS: This retrospective, single-institution study included 154 patients who underwent radical hysterectomy for early-stage squamous cell cervical cancer between 2007 and 2017. Tumor samples from 154 patients were available for L1CAM analysis by immunohistochemistry. Among all patients, radical abdominal hysterectomy was performed in 144 cases. RESULTS: L1CAM expression was positive in 24 tumors (15.6%) of the whole group. In relation to the grade of differentiation and the presence of lymphovascular invasion, L1CAM expression did not show an association (p=0.154 and p=0.306, respectively). The disease-free interval and overall survival also did not significantly differ between L1CAM-positive and L1CAM-negative cases (p=0.427 and p=0.240, respectively). For histopathological characteristics, L1CAM-positive cases had a significantly higher median tumor size (p=0.015). Even in the selected group of 115 cases without nodal infiltration, L1CAM status had no effect on the relapse rate during follow-up. CONCLUSION: Our study did not confirm the results of previous studies showing L1CAM expression to be a negative prognostic factor in cervical cancer. In our study, increased L1CAM expression in early-stage squamous-cell cervical cancer was not associated with adverse prognosis regarding disease recurrence, disease-free survival, nor overall survival. L1CAM expression was correlated only with the size of the tumor.


Asunto(s)
Carcinoma de Células Escamosas , Molécula L1 de Adhesión de Célula Nerviosa , Neoplasias del Cuello Uterino , Femenino , Humanos , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/análisis , Pronóstico , Estudios Retrospectivos , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
17.
Gene ; 881: 147643, 2023 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-37453721

RESUMEN

Hirschsprung disease (HSCR) is a congenital disorder of functional bowel obstruction due to the absence of enteric ganglia in distal bowel. Different L1cam variants were reportedly associated with L1cam syndrome and HSCR, whose phenotypes lacked predictable relevance to their genotypes. Using next-generation sequencing (NGS), we found an L1CAM de novo frameshift mutation in a female with mild hydrocephalus and skip-type HSCR. A nearly identical L1cam variant was introduced into FVB/NJ mice via the CRISPR-EZ method. A silent mutation was created via ssODN to gain an artificial Ncol restriction enzyme site for easier genotyping. Six L1cam protein-coding alternative transcripts were quantitatively measured. Immunofluorescence staining with polyclonal and monoclonal L1cam antibodies was used to characterize L1cam isoform proteins in enteric ganglia. Fifteen mice, seven males and eight females, generated via CRISPR-EZ, were confirmed to carry the L1cam frameshift variant, resulting in a premature stop codon. There was no prominent hydrocephalus nor HSCR-like presentation in these mice, but male infertility was noticed after observation for three generations in a total of 176 mice. Full-length L1cam transcripts were detected at a very low level in the intestinal tissues and almost none in the brain of these mice. Alternative shorter transcripts encoding the extracellular domains were overexpressed in the intestine of L1cam knockdown mice. Immunofluorescence confirmed no fulllength L1cam protein in enteric ganglia. These shorter L1cam isoform proteins might play a role in protecting L1cam knockdown mice from HSCR.


Asunto(s)
Enfermedad de Hirschsprung , Hidrocefalia , Molécula L1 de Adhesión de Célula Nerviosa , Animales , Femenino , Masculino , Ratones , Enfermedad de Hirschsprung/genética , Hidrocefalia/genética , Intestinos , Molécula L1 de Adhesión de Célula Nerviosa/genética , Isoformas de Proteínas
18.
Mol Genet Genomic Med ; 11(11): e2253, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37489051

RESUMEN

BACKGROUND: The L1 cell adhesion molecule (L1CAM, OMIM 308840) gene is primarily expressed in the nervous system and encodes the L1 adhesion molecule protein. Variations in L1CAM cause a wide spectrum of X-linked neurological disorders summarized as the L1 syndrome. METHODS: We report a 29-year-old pregnant woman who experienced multiple adverse pregnancy outcomes due to recurrent fetal hydrocephalus with an X-linked recessive inheritance. Genomic DNA was extracted from the third aborted male fetus and analyzed via trio whole-exome sequencing (WES). Total RNA was isolated from the pregnant woman to assess splicing variation at the mRNA level, and amniotic fluid was extracted from the woman for prenatal diagnosis on her fourth fetus. RESULTS: All four male fetuses were affected by severe hydrocephalus. We identified a maternally derived hemizygous splicing variation NM_000425.5:[c.3046 + 1G > A]; NP_000416.1 p.(Gly1016AspfsTer6) (chrX:153130275) in Intron 22 of the L1CAM. This variation disrupts the donor splice site and causes the retention of Intron 22, which results in frame shift and a premature termination codon at position 1021 with the ability to produce a truncated protein without the fifth fibronectin-repeat III, transmembrane, and cytoplasmic domains or to induce the degradation of mRNAs by nonsense-mediated mRNA decay. The same hemizygous variant was also detected in the amniocytes. CONCLUSION: This report enhances our knowledge of genetic and phenotypic characteristics of X-linked fetal hydrocephalus, providing a new genetic basis for prenatal diagnosis and pre-implantation prenatal diagnosis.


Asunto(s)
Hidrocefalia , Molécula L1 de Adhesión de Célula Nerviosa , Adulto , Femenino , Humanos , Masculino , Embarazo , Líquido Amniótico , Hidrocefalia/genética , Mutación , Molécula L1 de Adhesión de Célula Nerviosa/genética , ARN Mensajero/genética
19.
Sci Rep ; 13(1): 9243, 2023 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-37286800

RESUMEN

The Parkinson's disease (PD) associated protein, alpha-synuclein (α-syn/SNCA), is highly expressed in aggressive melanomas. The goal of this study was to reveal possible mechanism(s) of α-syn involvement in melanoma pathogenesis. Herein, we asked whether α-syn modulates the expression of the pro-oncogenic adhesion molecules L1CAM and N-cadherin. We used two human melanoma cell lines (SK-MEL-28, SK-MEL-29), SNCA-knockout (KO) clones, and two human SH-SY5Y neuroblastoma cell lines. In the melanoma lines, loss of α-syn expression resulted in significant decreases in the expression of L1CAM and N-cadherin and concomitant significant decreases in motility. On average, there was a 75% reduction in motility in the four SNCA-KOs tested compared to control cells. Strikingly, comparing neuroblastoma SH-SY5Y cells that have no detectable α-syn to SH-SY5Y cells that stably express α-syn (SH/+αS), we found that expressing α-syn increased L1CAM and single-cell motility by 54% and 597%, respectively. The reduction in L1CAM level in SNCA-KO clones was not due to a transcriptional effect, rather we found that L1CAM is more efficiently degraded in the lysosome in SNCA-KO clones than in control cells. We propose that α-syn is pro-survival to melanoma (and possibly neuroblastoma) because it promotes the intracellular trafficking of L1CAM to the plasma membrane.


Asunto(s)
Melanoma , Molécula L1 de Adhesión de Célula Nerviosa , Neuroblastoma , Humanos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Cadherinas , Melanoma/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neuroblastoma/patología
20.
Oncogene ; 42(27): 2166-2182, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37221223

RESUMEN

Due to the complexity and heterogeneity of breast cancer, the therapeutic effects of breast cancer treatment vary between subtypes. Breast cancer subtypes are classified based on the presence of molecular markers for estrogen or progesterone receptors and human epidermal growth factor 2. Thus, novel, comprehensive, and precise molecular indicators in breast carcinogenesis are urgently needed. Here, we report that ZNF133, a zinc-finger protein, is negatively associated with poor survival and advanced pathological staging of breast carcinomas. Moreover, ZNF133 is a transcription repressor physically associated with the KAP1 complex. It transcriptionally represses a cohort of genes, including L1CAM, that are critically involved in cell proliferation and motility. We also demonstrate that the ZNF133/KAP1 complex inhibits the proliferation and invasion of breast cancer cells in vitro and suppresses breast cancer growth and metastasis in vivo by dampening the transcription of L1CAM. Taken together, the findings of our study confirm the value of ZNF133 and L1CAM levels in the diagnosis and prognosis of breast cancer, contribute to a deeper understanding of the regulation mechanism of ZNF133 for the first time, and provide a new therapeutic strategy and precise intervention target for breast cancer.


Asunto(s)
Neoplasias de la Mama , Molécula L1 de Adhesión de Célula Nerviosa , Humanos , Femenino , Molécula L1 de Adhesión de Célula Nerviosa/genética , Invasividad Neoplásica , Proliferación Celular/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica , Línea Celular Tumoral , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA