Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Neurosci ; 42(39): 7355-7369, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36481742

RESUMEN

Interstitial collateral branching of axons is a critical component in the development of functional neural circuits. Axon collateral branches are established through a series of cellular processes initiated by the development of a specialized, focal F-actin network in axons. The formation, maintenance and remodeling of this F-actin patch is critical for the initiation of axonal protrusions that are subsequently consolidated to form a collateral branch. However, the mechanisms regulating F-actin patch dynamics are poorly understood. Fmn2 is a formin family member implicated in multiple neurodevelopmental disorders. We find that Fmn2 regulates the initiation of axon collateral protrusions in chick spinal neurons and in zebrafish motor neurons. Fmn2 localizes to the protrusion-initiating axonal F-actin patches and regulates the lifetime and size of these F-actin networks. The F-actin nucleation activity of Fmn2 is necessary for F-actin patch stability but not for initiating patch formation. We show that Fmn2 insulates the F-actin patches from disassembly by the actin-depolymerizing factor, ADF, and promotes long-lived, larger patches that are competent to initiate axonal protrusions. The regulation of axonal branching can contribute to the neurodevelopmental pathologies associated with Fmn2 and the dynamic antagonism between Fmn2 and ADF may represent a general mechanism of formin-dependent protection of Arp2/3-initiated F-actin networks from disassembly.SIGNIFICANCE STATEMENT Axonal branching is a key process in the development of functional circuits and neural plasticity. Axon collateral branching is initiated by the elaboration of F-actin filaments from discrete axonal F-actin networks. We show that the neurodevelopmental disorder-associated formin, Fmn2, is a critical regulator of axon collateral branching. Fmn2 localizes to the collateral branch-inducing F-actin patches in axons and regulates the stability of these actin networks. The F-actin nucleation activity of Fmn2 protects the patches from ADF-mediated disassembly. Opposing activities of Fmn2 and ADF exert a dynamic regulatory control on axon collateral branch initiation and may underly the neurodevelopmental defects associated with Fmn2.


Asunto(s)
Actinas , Pez Cebra , Animales , Citoesqueleto de Actina
2.
J Cell Sci ; 134(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34313311

RESUMEN

Dynamic co-regulation of the actin and microtubule subsystems enables the highly precise and adaptive remodelling of the cytoskeleton necessary for critical cellular processes, such as axonal pathfinding. The modes and mediators of this interpolymer crosstalk, however, are inadequately understood. We identify Fmn2, a non-diaphanous-related formin associated with cognitive disabilities, as a novel regulator of cooperative actin-microtubule remodelling in growth cones of both chick and zebrafish neurons. We show that Fmn2 stabilizes microtubules in the growth cones of cultured spinal neurons and in vivo. Super-resolution imaging revealed that Fmn2 facilitates guidance of exploratory microtubules along actin bundles into the chemosensory filopodia. Using live imaging, biochemistry and single-molecule assays, we show that a C-terminal domain in Fmn2 is necessary for the dynamic association between microtubules and actin filaments. In the absence of the cross-bridging function of Fmn2, filopodial capture of microtubules is compromised, resulting in destabilized filopodial protrusions and deficits in growth cone chemotaxis. Our results uncover a critical function for Fmn2 in actin-microtubule crosstalk in neurons and demonstrate that the modulation of microtubule dynamics via associations with F-actin is central to directional motility.


Asunto(s)
Actinas , Quimiotaxis , Forminas/genética , Conos de Crecimiento , Neuronas/citología , Citoesqueleto de Actina , Animales , Axones , Pollos , Microtúbulos , Pez Cebra
3.
Fetal Pediatr Pathol ; 40(6): 702-706, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32162566

RESUMEN

Introduction Genetic causes of the intellectual disability Nonsyndromic Autosomal-Recessive Intellectual Disability Syndrome (MRT47, MIM 616193) are mutations in the recently described FMN2 (formin 2 gene). Case report: A boy with intellectual disability had a novel homozygous nonsense mutation (c.2245C > T/p.Gln749*) leading to a premature stop codon in exon 6 of the FMN2 (NM_001305424) gene detected by Clinical Exome Sequencing (CES). Conclusion: Clinical features of a patient with a novel nonsense FMN2 mutation is presented. We urge the change in the OMIM nomenclature from Mental Retardation, Autosomal Recessive 47 (MRT47, MIM 616193) to 'Nonsyndromic Autosomal-Recessive Intellectual Disability Syndrome'.


Asunto(s)
Codón sin Sentido , Discapacidad Intelectual , Niño , Genes Recesivos , Homocigoto , Humanos , Discapacidad Intelectual/genética , Masculino , Mutación , Linaje
4.
J Gene Med ; 19(6-7)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27556228

RESUMEN

BACKGROUND: In the present study, we explored the functional roles of microRNA-144 (miR-144) upregulation and downregulation in human acute lymphoblastic leukemia (ALL). METHODS: Gene expression of miR-144 was examined using the quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) in both ALL cell lines and T-leukemic cells of ALL patients. In ALL cell lines Molt-3 and Jurkat cells, miR-144 was either upregulated or downregulated through lentiviral transduction. The subsequent effects of miR-144 upregulation or downregulation on ALL proliferation, cell-cycle transition and in vivo xenograft were assessed. A possible downstream target of miR-144, human formin-2 (FMN2), was assessed by a dual-luciferase activity assay, qRT-PCR and western blotting. FMN2 was then overexpressed in miR-144-upregulated Molt-3 and Jurkat cells to determine its effect on miR-144 induced tumor suppression on ALL proliferation and cell-cycle transition. RESULTS: MiR-144 was significantly downregulated in both ALL cell lines and T-leukemic cells of ALL patients. Lentiviral transfection successfully induced miR-144 upregulation or downregulation in Molt-3 and Jurkat cells without affecting cell viability. Functional assays demonstrated that miR-144 upregulation suppressed ALL proliferation and cell-cycle transition in vitro, as well as the growth of Jurkat xenograft in vivo, whereas miR-144 downregulation had no functional effect on ALL development. Multiple approaches confirmed that FMN2 was the downstream target of miR-144 in ALL. Finally, overexpressing FMN2 reversed the inhibitory effects of miR-144-upregulation on ALL proliferation and cell-cycle transition. CONCLUSIONS: MiR-144 functions as a tumor suppressor in ALL, very likely through the inverse regulation of its target gene FMN2.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Forminas , Humanos , Células Jurkat , MicroARNs/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo
5.
Appl Biochem Biotechnol ; 195(12): 7322-7337, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36995659

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract with high incidence and mortality. Exosomal circular RNA (circRNA) has been shown to be associated with the malignant progression of cancers, including CRC. Circ_0005100 (named as circ_FMN2) has been shown to promote CRC cell proliferation and migration. However, whether exosomal circ_FMN2 participated in CRC progression remains unclear. METHODS: Exosomes were isolated from the serum of CRC patients and then identified using transmission electron microscope. Western blot assay was used to test the protein levels of exosome markers, proliferation-related marker, metastasis-related markers and musashi-1 (MSI1). The expression levels of circ_FMN2, microRNA (miR)-338-3p and MSI1 were detected by qPCR. Flow cytometry, colony formation assay, MTT assay, and transwell assay were employed to measure cell cycle, apoptosis, colony formation ability, viability, migration and invasion. Dual-luciferase reporter assay was performed to assess the interaction between miR-338-3p and circ_FMN2 or MSI1. BALB/c nude mice was used to conduct animal experiments. RESULTS: Circ_FMN2 was overexpressed in the exosomes of CRC patient's serums and CRC cells. Overexpressed exosomal circ_FMN2 could promote CRC cell proliferation, metastasis, and suppress apoptosis. Circ_FMN2 acted as miR-338-3p sponge. MiR-338-3p overexpression reversed the promotion effect of circ_FMN2 on CRC progression. MSI1 was found to be a target of miR-338-3p, and its overexpression revoked the inhibitory effect of miR-338-3p on CRC progression. Furthermore, exosomal circ_FMN2 overexpression also could facilitate CRC tumor growth in vivo. CONCLUSION: Exosomal circ_FMN2 accelerated CRC progression through miR-338-3p/MSI1 axis, revealing that exosomal circ_FMN2 might be a target for CRC treatment.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Animales , Humanos , Ratones , Apoptosis , Vendajes , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Colorrectales/genética , Ratones Desnudos , MicroARNs/genética , Proteínas del Tejido Nervioso , Proteínas de Unión al ARN/genética
6.
J Ovarian Res ; 15(1): 31, 2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35227295

RESUMEN

BACKGROUND: Premature ovarian insufficiency (POI) plagues 1% of women under 40, while quite a few remain an unknown cause. The development of sequencing has helped find pathogenic genes and reveal the relationship between DNA repair and ovarian reserve. Through the exome sequencing, our study targets screening out the possible POI pathogenic gene and variants in a Chinese family and 20 sporadic POI patients, preliminarily exploring the functional impact and finding out potential linkages between the gene and POI. RESULTS: The whole exome sequencing suggested a novel FMN2 heterozygous variant c.1949C > T (p.Ser650Leu) carried by all three patients in a Chinese family and another c.1967G > A(p.Arg656His) variant in a sporadic case. Since no FMN2 missense mutation is reported for causing human POI, we preliminarily assessed p.Ser650Leu variant via cross-species alignment and 3D modeling and found it possibly deleterious. A series of functional evidence was consistent with our hypothesis. We proved the expression of FMN2 in different stages of oocytes and observed a statistical difference of chromosomal breakages between the POI patient carrying p.Arg656His variant and the health control (p = 0.0013). Western Blot also suggested a decrease in FMN2 and P21 in the mutant type and an associated increase in H2AX. The p.Arg656His variant with an extremely low frequency also indicated that the gene FMN2 might play an essential role in the genetic etiology of POI. To the best of our knowledge, this is the first POI report on missense variants of FMN2. CONCLUSION: This finding indicates a novel gene possibly related to POI and sheds lights on the study of FMN2.


Asunto(s)
Forminas/genética , Forminas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Insuficiencia Ovárica Primaria/genética , Adulto , Reparación del ADN/genética , Femenino , Feto/metabolismo , Heterocigoto , Histonas/sangre , Humanos , Linfocitos/metabolismo , Estructura Molecular , Mutación Missense , Ovario/metabolismo , Linaje , Insuficiencia Ovárica Primaria/sangre , Secuenciación del Exoma
7.
eNeuro ; 8(4)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34193512

RESUMEN

The formin family member Fmn2 is a neuronally enriched cytoskeletal remodeling protein conserved across vertebrates. Recent studies have implicated Fmn2 in neurodevelopmental disorders, including sensory processing dysfunction and intellectual disability in humans. Cellular characterization of Fmn2 in primary neuronal cultures has identified its function in the regulation of cell-substrate adhesion and consequently growth cone translocation. However, the role of Fmn2 in the development of neural circuits in vivo, and its impact on associated behaviors have not been tested. Using automated analysis of behavior and systematic investigation of the associated circuitry, we uncover the role of Fmn2b in zebrafish neural circuit development. As reported in other vertebrates, the zebrafish ortholog of Fmn2 is also enriched in the developing zebrafish nervous system. We find that Fmn2b is required for the development of an excitatory interneuron pathway, the spiral fiber neuron, which is an essential circuit component in the regulation of the Mauthner cell (M-cell)-mediated acoustic startle response. Consistent with the loss of the spiral fiber neurons tracts, high-speed video recording revealed a reduction in the short latency escape events while responsiveness to the stimuli was unaffected. Taken together, this study provides evidence for a circuit-specific requirement of Fmn2b in eliciting an essential behavior in zebrafish. Our findings underscore the importance of Fmn2 in neural development across vertebrate lineages and highlight zebrafish models in understanding neurodevelopmental disorders.


Asunto(s)
Forminas , Reflejo de Sobresalto , Proteínas de Pez Cebra , Pez Cebra , Acústica , Animales , Humanos , Interneuronas
8.
Neuroscience ; 448: 160-171, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33002558

RESUMEN

Growth cone-mediated axonal outgrowth and accurate synaptic targeting are central to brain morphogenesis. Translocation of the growth cone necessitates mechanochemical regulation of cell-extracellular matrix interactions and the generation of propulsive traction forces onto the growth environment. However, the molecular mechanisms subserving force generation by growth cones remain poorly characterized. The formin family member, Fmn2, has been identified earlier as a regulator of growth cone motility. Here, we explore the mechanisms underlying Fmn2 function in the growth cone. Evaluation of multiple components of the adhesion complexes suggests that Fmn2 regulates point contact stability. Analysis of F-actin retrograde flow reveals that Fmn2 functions as a clutch molecule and mediates the coupling of the actin cytoskeleton to the growth substrate, via point contact adhesion complexes. Using traction force microscopy, we show that the Fmn2-mediated clutch function is necessary for the generation of traction stresses by neurons. Our findings suggest that Fmn2, a protein associated with neurodevelopmental and neurodegenerative disorders, is a key regulator of a molecular clutch activity and consequently motility of neuronal growth cones.


Asunto(s)
Forminas/genética , Conos de Crecimiento , Proteínas Nucleares/genética , Actinas , Movimiento Celular , Neuronas
9.
BMC Med Genomics ; 11(1): 50, 2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29801487

RESUMEN

BACKGROUND: In children with sensory processing dysfunction (SPD), who do not meet criteria for autism spectrum disorder (ASD) or intellectual disability, the contribution of de novo pathogenic mutation in neurodevelopmental genes is unknown and in need of investigation. We hypothesize that children with SPD may have pathogenic variants in genes that have been identified as causing other neurodevelopmental disorders including ASD. This genetic information may provide important insight into the etiology of sensory processing dysfunction and guide clinical evaluation and care. METHODS: Eleven community-recruited trios (children with isolated SPD and both biological parents) underwent WES to identify candidate de novo variants and inherited rare single nucleotide variants (rSNV) in genes previously associated with ASD. Gene enrichment in these children and their parents for transmitted and non-transmitted mutation burden was calculated. A comparison analysis to assess for enriched rSNV burden was then performed in 2377 children with ASD and their families from the Simons Simplex Collection. RESULTS: Of the children with SPD, 2/11 (18%), were identified as having a de novo loss of function or missense mutation in genes previously reported as causative for neurodevelopmental disorders (MBD5 and FMN2). We also found that the parents of children with SPD have significant enrichment of pathogenic rSNV burden in high-risk ASD candidate genes that are inherited by their affected children. Using the same approach, we confirmed enrichment of rSNV burden in a large cohort of children with autism and their parents but not unaffected siblings. CONCLUSIONS: Our findings suggest that SPD, like autism, has a genetic basis that includes both de novo single gene mutations as well as an accumulated burden of rare inherited variants from their parents.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Mutación , Sensación/genética , Trastorno del Espectro Autista/psicología , Niño , Estudios de Cohortes , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Padres , Hermanos
10.
Eur J Cell Biol ; 93(5-6): 225-37, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24345451

RESUMEN

Spir proteins nucleate actin filaments at vesicle membranes and facilitate intracellular transport processes. The mammalian genome encodes two Spir proteins, namely Spir-1 and Spir-2. While the mouse spir-2 gene has a rather broad expression pattern, high levels of spir-1 expression are restricted to the nervous system, oocytes, and testis. Spir-1 mutant mice generated by a gene trap method have been employed to address Spir-1 function during mouse development and in adult mouse tissues, with a specific emphasis on viability, reproduction, and the nervous system. The gene trap cassette disrupts Spir-1 expression between the N-terminal KIND domain and the WH2 domain cluster. Spir-1 mutant mice are viable and were born in a Mendelian ratio. In accordance with the redundant function of Spir-1 and Spir-2 in oocyte maturation, spir-1 mutant mice are fertile. The overall brain anatomy of spir-1 mutant mice is not altered and visual and motor functions of the mice remain normal. Microscopic analysis shows a slight reduction in the number of dendritic spines on cortical neurons. Detailed behavioral studies of the spir-1 mutant mice, however, unveiled a very specific and highly significant phenotype in terms of fear learning in male mice. In contextual and cued fear conditioning experiments the male spir-1 mutant mice display increased fear memory when compared to their control littermates. Our data point toward a particular function of the vesicle associated Spir-1 actin organizer in neuronal circuits determining fear behavior.


Asunto(s)
Actinas/metabolismo , Miedo/psicología , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Condicionamiento Clásico , Dendritas/metabolismo , Espinas Dendríticas/ultraestructura , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas de Microfilamentos/metabolismo , Actividad Motora , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Vesículas Transportadoras/genética , Vesículas Transportadoras/metabolismo , Percepción Visual
11.
Eur J Med Genet ; 56(12): 686-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24161494

RESUMEN

We present the case of a child with mild non-syndromic intellectual disability in whom array genomic hybridization revealed a de novo heterozygous deletion involving only one gene, FMN2. FMN2 encodes FORMIN-2, a member of the formin homology family, which is primarily expressed in the developing and mature brain, and has an important role in cytoskeletal organization and actin nucleation. A heterozygous deletion of FMN2 along with 2 other genes has been recently reported in a boy with non-syndromic intellectual disability. This report provides further support for the important role of FMN2 in brain development and cognition.


Asunto(s)
Eliminación de Gen , Discapacidad Intelectual/genética , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Niño , Femenino , Forminas , Heterocigoto , Humanos , Discapacidad Intelectual/diagnóstico
12.
Cell Cycle ; 12(15): 2348-54, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23839046

RESUMEN

We have identified the human FMN2 gene as a novel target regulated by induction of p14ARF and by multiple other stress responses, including DNA damage and hypoxia, which have in common activation of cell cycle arrest. We showed that increased expression of the FMN2 gene following p14ARF induction is caused, at the transcriptional level, by relief of repression by RelA and E2F1, which, under non-induced conditions, bind the FMN2 promoter. Increased FMN2 protein levels promote cell cycle arrest by inhibiting the degradation of p21, and our data show that control of p21 stability is a key part of the mechanism that regulates p21 induction. Consistent with this model, we have shown that transient expression of exogenous FMN2 protein alone is sufficient to increase p21 protein levels in cells, without altering p21 mRNA levels. Here, we provide additional evidence for the role of the N terminus of FMN2 as being the important domain required for p21 stability. In addition, we also investigate the role of RelA's threonine 505 residue in the control of FMN2. Our results identify FMN2 as a crucial protein involved in the control of p21.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Microfilamentos/fisiología , Proteínas Nucleares/fisiología , Animales , Hipoxia de la Célula , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Daño del ADN , Forminas , Regulación de la Expresión Génica , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteolisis , Transducción de Señal , Factor de Transcripción ReIA/metabolismo
13.
Artículo en Zh | WPRIM | ID: wpr-812033

RESUMEN

@# Objective: To investigate the expression of Formin-2(FMN2)protein in gastric cancer tissues and its correlation to clinicopathological features of gastric cancer patients, as well to explore its effect on the proliferation of gastric adenocarcinoma AGS cells. Methods: 84 cases of gastric adenocarcinoma tissues and corresponding para-cancerous tissues were surgically collected from patients treated in the First Affiliated Hospital of Air Force Military Medical University from September 2015 to September 2017. The expression of FMN2 in gastric adenocarcinoma tissues was detected by immunohistochemical staining and analyzed with RNA-Seq data-sets GEPIA. The relationship between FMN2 protein expression in gastric adenocarcinoma tissues and its clinicopathological features was also explored. MTT assay was used to detect the effect of FMN2 onAGS cell proliferation activity, and Western blotting was used to detect the effect of FMN2 on the expression of apoptosis-related protein caspase-3 in AGS cells. Results: The expression level of FMN2 in gastric adenocarcinoma tissues was significantly lower than that in matched adjacent tissues and the expression level of FMN2 was closely related to the TNM stage and differentiation of gastric adenocarcinoma (all P<0.05). Compared toAGS control group, the proliferation activity of AGS/FMN2 was significantly decreased and the expression of apoptosis-related gene Caspase-3 was markedly increased (all P<0.05). Conclusion: The expression of FMN2 was significantly decreased in gastric adenocarcinoma tissues and its low expression is closely related to the degree of tumor differentiation and clinical TNM stage. Moreover, FMN2 over-expression significantly decreased the proliferation of AGS cells. FMN2 may function as independent risk factor for the prognosis of gastric adenocarcinoma, which may provide new ideas for the treatment of gastric adenocarcinoma.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda