Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
J Neurosci ; 44(8)2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38267260

RESUMO

The inner ear sensory neurons play a pivotal role in auditory processing and balance control. Though significant progresses have been made, the underlying mechanisms controlling the differentiation and survival of the inner ear sensory neurons remain largely unknown. During development, ISL1 and POU4F transcription factors are co-expressed and are required for terminal differentiation, pathfinding, axon outgrowth and the survival of neurons in the central and peripheral nervous systems. However, little is understood about their functional relationship and regulatory mechanism in neural development. Here, we have knocked out Isl1 or Pou4f1 or both in mice of both sexes. In the absence of Isl1, the differentiation of cochleovestibular ganglion (CVG) neurons is disturbed and with that Isl1-deficient CVG neurons display defects in migration and axon pathfinding. Compound deletion of Isl1 and Pou4f1 causes a delay in CVG differentiation and results in a more severe CVG defect with a loss of nearly all of spiral ganglion neurons (SGNs). Moreover, ISL1 and POU4F1 interact directly in developing CVG neurons and act cooperatively as well as independently in regulating the expression of unique sets of CVG-specific genes crucial for CVG development and survival by binding to the cis-regulatory elements including the promoters of Fgf10, Pou4f2, and Epha5 and enhancers of Eya1 and Ntng2 These findings demonstrate that Isl1 and Pou4f1 are indispensable for CVG development and maintenance by acting epistatically to regulate genes essential for CVG development.


Assuntos
Orelha Interna , Regulação da Expressão Gênica no Desenvolvimento , Animais , Feminino , Masculino , Camundongos , Gânglios/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Células Receptoras Sensoriais/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Genesis ; 57(10): e23328, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31313880

RESUMO

LHX4 is a LIM-homeodomain transcription factor essential for the development of spinal cord and pituitary gland. Mice with homozygous Lhx4-null mutation suffer early postnatal death from lung defect. In this study, to facilitate the research on Lhx4 function, we designed a targeting construct to generate two novel Lhx4 mouse lines: Lhx4 loxP conditional knockout and Lhx4 tdT reporter knock-in mice. Lhx4 tdT/+ , Lhx4 loxP/+ , and Lhx4 loxP/loxP were viable, fertile, and did not display any gross abnormalities. By breeding Lhx4 loxP line with Cre-expressing mice, the Exon 3 of Lhx4 was efficiently removed, resulting in a shift in the reading frame and the inactivation of Lhx4. The expression of tdTomato knock-in reporter recapitulated the endogenous LHX4 expression and was detected in the retina, spinal cord, pituitary gland, and hindbrain of Lhx4 tdT mice. Thus, Lhx4 tdT and Lhx4 loxP mouse lines provide valuable tools for unraveling the tissue-specific role of Lhx4 at postnatal stages in mice.


Assuntos
Genes Reporter , Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Animais , Técnicas de Introdução de Genes , Integrases/genética , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutagênese Insercional , Fatores de Transcrição/metabolismo , Proteína Vermelha Fluorescente
3.
Acta Derm Venereol ; 99(12): 1110-1115, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31513274

RESUMO

Nail-patella syndrome is an autosomal dominant disorder characterized by nail dysplasia and skeletal anomaly. Some patients have been shown to have ultrastructural abnormalities of the glomerular basement membrane that result in nephrosis. However, little has been reported on the epidermal basement membrane in this condition. This paper reports 2 families with nail-patella syndrome. Direct sequencing analysis of LMX1B revealed that family 1 and family 2 were heterozygous for the mutations c.140-1G>C and c.326+1G>C, respectively. To evaluate the epidermal basement membrane zone, ultrastructural and immunohistochemical analyses were performed using skin specimens obtained from the dorsal thumb. Electron microscopy showed intact hemidesmosomes, lamina lucida, lamina densa, and anchoring fibrils. Immunofluorescence studies with antibodies against components of the epidermal basement membrane zone revealed a normal expression pattern among the components, including type IV collagen. These data suggest that nail dysplasia in patients with nail-patella syndrome is not caused by structural abnormalities of the epidermal basement membrane.


Assuntos
Membrana Basal/química , Membrana Basal/ultraestrutura , Colágeno Tipo IV/análise , Epiderme/química , Epiderme/ultraestrutura , Imunofluorescência , Microscopia Eletrônica de Transmissão , Síndrome da Unha-Patela/diagnóstico , Biomarcadores/análise , Criança , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Lactente , Proteínas com Homeodomínio LIM/genética , Masculino , Mutação , Síndrome da Unha-Patela/genética , Síndrome da Unha-Patela/metabolismo , Síndrome da Unha-Patela/patologia , Fenótipo , Valor Preditivo dos Testes , Fatores de Transcrição/genética
4.
Genesis ; 56(4): e23098, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29508544

RESUMO

LHX3, a LIM-homeodomain transcription factor, is broadly expressed in the developing pituitary, spinal cord, medulla, retina and inner ear, and plays essential roles during embryonic development. Mice with homozygous Lhx3 null mutation exhibit failure in the formation of pituitary gland and die perinatally. To facilitate the functional study of Lhx3 in mice, we engineered and characterized two novel Lhx3 mouse strains: Lhx3GFP reporter knock-in and Lhx3loxP conditional knockout mice. Coimmunolabeling of LHX3 and GFP shows that the expression pattern of the knock-in GFP reporter recapitulates that of endogenous LHX3 in cochlea, vestibule, retina, and spinal cord. By crossing Lhx3loxP mice with the ubiquitous CMV-Cre mice, we have demonstrated a high efficiency of Cre recombinase-mediated removal of exons 3 to 5 of Lhx3, which encode the second LIM-domain and the HD domain of LHX3, resulting global knockout of Lhx3. Thus, Lhx3GFP and Lhx3loxP mice serve as valuable genetic tools to dissect the tissue-specific roles of Lhx3 at late-gestation and postnatal stages in mice.


Assuntos
Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Éxons , Regulação da Expressão Gênica no Desenvolvimento/genética , Engenharia Genética/métodos , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Mutação , Hipófise/metabolismo , Hipófise/fisiologia
5.
Proc Natl Acad Sci U S A ; 110(28): 11445-50, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23798382

RESUMO

The motor neuron (MN)-hexamer complex consisting of LIM homeobox 3, Islet-1, and nuclear LIM interactor is a key determinant of motor neuron specification and differentiation. To gain insights into the transcriptional network in motor neuron development, we performed a genome-wide ChIP-sequencing analysis and found that the MN-hexamer directly regulates a wide array of motor neuron genes by binding to the HxRE (hexamer response element) shared among the target genes. Interestingly, STAT3-binding motif is highly enriched in the MN-hexamer-bound peaks in addition to the HxRE. We also found that a transcriptionally active form of STAT3 is expressed in embryonic motor neurons and that STAT3 associates with the MN-hexamer, enhancing the transcriptional activity of the MN-hexamer in an upstream signal-dependent manner. Correspondingly, STAT3 was needed for motor neuron differentiation in the developing spinal cord. Together, our studies uncover crucial gene regulatory mechanisms that couple MN-hexamer and STAT-activating extracellular signals to promote motor neuron differentiation in vertebrate spinal cord.


Assuntos
Diferenciação Celular/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Neurônios Motores/citologia , Fator de Transcrição STAT3/fisiologia , Animais , Elementos Facilitadores Genéticos , Genoma , Camundongos , Ligação Proteica , Fator de Transcrição STAT3/metabolismo
6.
Genesis ; 52(9): 827-832, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25112520

RESUMO

LHX9 is a LIM-homeodomain transcription factor essential for the development of gonads, spinal cord interneurons, and thalamic neurons to name a few. We recently reported the expression of LHX9 in retinal amacrine cells during development. In this study, we generated an Lhx9-GFPCreER(T) (2) (GCE) knock-in mouse line by knocking-in a GCE cassette at the Lhx9 locus, thus inactivating endogenous Lhx9. Lhx9(GCE) (/+) mice were viable, fertile, and displayed no overt phenotypical characteristics. Lhx9(GCE) (/) (GCE) mice were all phenotypically female, smaller in size, viable, but infertile. The specificity and efficacy of the Lhx9-GCE mouse line was verified by crossing it to a Rosa26-tdTomato reporter mouse line, which reveals the Cre recombinase activities in retinal amacrine cells, developing limbs, testis, hippocampal neurons, thalamic neurons, and cerebellar neurons. Taken together, the Lhx9-GCE mouse line could serve as a beneficial tool for lineage tracing and gene manipulation experiments. genesis


Assuntos
Proteínas com Homeodomínio LIM/genética , Fatores de Transcrição/genética , Células Amácrinas/citologia , Células Amácrinas/metabolismo , Animais , Extremidades/embriologia , Extremidades/crescimento & desenvolvimento , Feminino , Técnicas de Introdução de Genes , Loci Gênicos , Hipocampo/embriologia , Hipocampo/crescimento & desenvolvimento , Integrases/genética , Integrases/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Neurônios/microbiologia , Retina/citologia , Retina/embriologia , Retina/crescimento & desenvolvimento , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/metabolismo
7.
J Biol Chem ; 288(30): 21924-35, 2013 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-23750000

RESUMO

Islet 1 (Isl1) is a transcription factor of the LIM-homeodomain (LIM-HD) protein family and is essential for many developmental processes. LIM-HD proteins all contain two protein-interacting LIM domains, a DNA-binding homeodomain (HD), and a C-terminal region. In Isl1, the C-terminal region also contains the LIM homeobox 3 (Lhx3)-binding domain (LBD), which interacts with the LIM domains of Lhx3. The LIM domains of Isl1 have been implicated in inhibition of DNA binding potentially through an intramolecular interaction with or close to the HD. Here we investigate the LBD as a candidate intramolecular interaction domain. Competitive yeast-two hybrid experiments indicate that the LIM domains and LBD from Isl1 can interact with apparently low affinity, consistent with no detection of an intermolecular interaction in the same system. Nuclear magnetic resonance studies show that the interaction is specific, whereas substitution of the LBD with peptides of the same amino acid composition but different sequence is not specific. We solved the crystal structure of a similar but higher affinity complex between the LIM domains of Isl1 and the LIM interaction domain from the LIM-HD cofactor protein LIM domain-binding protein 1 (Ldb1) and used these coordinates to generate a homology model of the intramolecular interaction that indicates poorer complementarity for the weak intramolecular interaction. The intramolecular interaction in Isl1 may provide protection against aggregation, minimize unproductive DNA binding, and facilitate cofactor exchange within the cell.


Assuntos
Proteínas com Homeodomínio LIM/química , Proteínas com Homeodomínio LIM/metabolismo , Estrutura Terciária de Proteína , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Cristalografia por Raios X , Proteínas com Homeodomínio LIM/genética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Ligação Proteica , Saccharomyces cerevisiae/genética , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido
8.
Cereb Cortex ; 23(8): 1811-23, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22710612

RESUMO

Deletion of LIM homeodomain transcription factor-encoding Lhx6 gene in mice results in defective tangential migration of cortical interneurons and failure of differentiation of the somatostatin (Sst)- and parvalbumin (Pva)-expressing subtypes. Here, we characterize a novel hypomorphic allele of Lhx6 and demonstrate that reduced activity of this locus leads to widespread differentiation defects in Sst(+) interneurons, but relatively minor and localized changes in Pva(+) interneurons. The reduction in the number of Sst-expressing cells was not associated with a loss of interneurons, because the migration and number of Lhx6-expressing interneurons and expression of characteristic molecular markers, such as calretinin or Neuropeptide Y, were not affected in Lhx6 hypomorphic mice. Consistent with a selective deficit in the differentiation of Sst(+) interneurons in the CA1 subfield of the hippocampus, we observed reduced expression of metabotropic Glutamate Receptor 1 in the stratum oriens and characteristic changes in dendritic inhibition, but normal inhibitory input onto the somatic compartment of CA1 pyramidal cells. Moreover, Lhx6 hypomorphs show behavioral, histological, and electroencephalographic signs of recurrent seizure activity, starting from early adulthood. These results demonstrate that Lhx6 plays an important role in the maturation of cortical interneurons and the formation of inhibitory circuits in the mammalian cortex.


Assuntos
Córtex Cerebral/fisiologia , Interneurônios/fisiologia , Proteínas com Homeodomínio LIM/fisiologia , Rede Nervosa/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fatores de Transcrição/fisiologia , Animais , Movimento Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Interneurônios/citologia , Proteínas com Homeodomínio LIM/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética
9.
Open Biol ; 13(12): 230327, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38086422

RESUMO

Adult planarians can regenerate the gut, eyes and even a functional brain. Proper identity and patterning of the newly formed structures require signals that guide and commit their adult stem cells. During embryogenesis, LIM-homeodomain (LIM-HD) transcription factors act in a combinatorial 'LIM code' to control cell fate determination and differentiation. However, our understanding about the role these genes play during regeneration and homeostasis is limited. Here, we report the full repertoire of LIM-HD genes in Schmidtea mediterranea. We found that lim homeobox (lhx) genes appear expressed in complementary patterns along the cephalic ganglia and digestive system of the planarian, with some of them being co-expressed in the same cell types. We have identified that Smed-islet1, -lhx1/5-1, -lhx2/9-3, -lhx6/8, -lmx1a/b-2 and -lmx1a/b-3 are essential to pattern and size the planarian brain as well as for correct regeneration of specific subpopulations of dopaminergic, serotonergic, GABAergic and cholinergic neurons, while Smed-lhx1/5.2 and -lhx2/9.2 are required for the proper expression of intestinal cell type markers, specifically the goblet subtype. LIM-HD are also involved in controlling axonal pathfinding (lhx6/8), axial patterning (islet1, lhx1/5-1, lmx1a/b-3), head/body proportions (islet2) and stem cell proliferation (lhx3/4, lhx2/9-3, lmx1a/b-2, lmx1a/b-3). Altogether, our results suggest that planarians might present a combinatorial LIM code that controls axial patterning and axonal growing and specifies distinct neuronal and intestinal cell identities.


Assuntos
Planárias , Fatores de Transcrição , Animais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Planárias/genética , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neurônios/metabolismo
10.
J Struct Biol X ; 5: 100043, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33458649

RESUMO

The roles of ISL1 and LHX3 in the development of spinal motor neurons have been well established. Whereas LHX3 triggers differentiation into interneurons, the additional expression of ISL1 in developing neuronal cells is sufficient to redirect their developmental trajectory towards spinal motor neurons. However, the underlying mechanism of this action by these transcription factors is less well understood. Here, we used electrophoretic mobility shift assays (EMSAs) and surface plasmon resonance (SPR) to probe the different DNA-binding behaviours of these two proteins, both alone and in complexes mimicking those found in developing neurons, and found that ISL1 shows markedly different binding properties to LHX3. We used small angle X-ray scattering (SAXS) to structurally characterise DNA-bound species containing ISL1 and LHX3. Taken together, these results have allowed us to develop a model of how these two DNA-binding modules coordinate to regulate gene expression and direct development of spinal motor neurons.

11.
Neuroscience ; 452: 247-264, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33246067

RESUMO

The inner ear, projections, and brainstem nuclei are essential components of the auditory and vestibular systems. It is believed that the evolution of complex systems depends on duplicated sets of genes. The contribution of duplicated genes to auditory or vestibular system development, however, is poorly understood. We describe that Lmx1a and Lmx1b, which originate from the invertebrate Lmx1b-like gene, redundantly regulate development of multiple essential components of the mammalian auditory/vestibular systems. Combined, but not individual, loss of Lmx1a/b eliminated the auditory inner ear organ of Corti (OC) and disrupted the spiral ganglion, which was preceded by a diminished expression of their critical regulator Pax2. Innervation of the remaining inner ear vestibular organs revealed unusual sizes or shapes and was more affected compared to Lmx1a/b single-gene mutants. Individual loss of Lmx1a/b genes did not disrupt brainstem auditory nuclei or inner ear central projections. Combined loss of Lmx1a/b, however, eliminated excitatory neurons in cochlear/vestibular nuclei, and also the expression of a master regulator Atoh1 in their progenitors in the lower rhombic lip (RL). Finally, in Lmx1a/b double mutants, vestibular afferents aberrantly projected to the roof plate. This phenotype was associated with altered expression of Wnt3a, a secreted ligand of the Wnt pathway that regulates pathfinding of inner ear projections. Thus, Lmx1a/b are redundantly required for the development of the mammalian inner ear, inner ear central projections, and cochlear/vestibular nuclei.


Assuntos
Órgão Espiral , Fatores de Transcrição , Animais , Cóclea , Proteínas com Homeodomínio LIM/genética , Gânglio Espiral da Cóclea , Fatores de Transcrição/genética
12.
Cell Rep ; 32(11): 108144, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32937137

RESUMO

Retinal bipolar cells (BCs) connect with photoreceptors and relay visual information to retinal ganglion cells (RGCs). Retina-specific deletion of Lhx4 in mice results in a visual defect resembling human congenital stationary night blindness. This visual dysfunction results from the absence of rod bipolar cells (RBCs) and the loss of selective rod-connecting cone bipolar cell (CBC) subtypes and AII amacrine cells (ACs). Inactivation of Lhx4 causes the apoptosis of BCs and cell fate switch from some BCs to ACs, whereas Lhx4 overexpression promotes BC genesis. Moreover, Lhx4 positively regulates Lhx3 expression to drive the fate choice of type 2 BCs over the GABAergic ACs. Lhx4 inactivation ablates Bhlhe23 expression, whereas overexpression of Bhlhe23 partially rescues RBC development in the absence of Lhx4. Thus, by acting upstream of Bhlhe23, Prdm8, Fezf2, Lhx3, and other BC genes, Lhx4, together with Isl1, could play essential roles in regulating the subtype-specific development of RBCs and CBCs.


Assuntos
Diferenciação Celular , Proteínas com Homeodomínio LIM/metabolismo , Células Bipolares da Retina/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Fatores de Transcrição/metabolismo , Envelhecimento/patologia , Células Amácrinas/metabolismo , Células Amácrinas/patologia , Animais , Apoptose , Eletrorretinografia , Proteínas com Homeodomínio LIM/deficiência , Camundongos , Visão Noturna , Células Bipolares da Retina/patologia , Células Fotorreceptoras Retinianas Cones/patologia , Fatores de Transcrição/deficiência , Transcriptoma/genética
13.
Vasc Endovascular Surg ; 54(2): 175-181, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31746280

RESUMO

Nail-patella syndrome (NPS) is a rare disorder characterized by abnormal development of ectodermal and mesodermal tissues. Classically, NPS presents as a triad of nail dysplasia, dysplastic patellae, and bony exostoses of the ilia known as "iliac horns." Apart from dermatological and skeletal abnormalities, patients may also have involvement of ophthalmologic and renal systems. The underlying molecular etiology in NPS is the mutation of LMX1B homeobox gene which results in loss of function of its protein also called LMX1B, a DNA-binding protein belonging to the larger LIM-homeodomain transcription factor family. Normal LMX1B gene and protein function are essential for dorsalization of the vertebrate limb bud, development of anterior eye structures, skull formation, and differentiation and migration of neurons in the central nervous system. We report a case of confirmed NPS presenting with congenital aplasia of the internal carotid artery and believe this is the first report of cerebrovascular developmental abnormality associated with NPS.


Assuntos
Artéria Carótida Interna/anormalidades , Malformações Vasculares do Sistema Nervoso Central/genética , Proteínas com Homeodomínio LIM/genética , Mutação , Síndrome da Unha-Patela/genética , Fatores de Transcrição/genética , Artéria Carótida Interna/diagnóstico por imagem , Malformações Vasculares do Sistema Nervoso Central/diagnóstico por imagem , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Síndrome da Unha-Patela/diagnóstico , Fenótipo
14.
Front Mol Neurosci ; 11: 271, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30127719

RESUMO

During mammalian retinal development, the multipotent progenitors differentiate into all classes of retinal cells under the delicate control of transcriptional factors. The deficiency of a transcription cofactor, the LIM-domain binding protein Ldb1, has been shown to cause proliferation and developmental defects in multiple tissues including cardiovascular, hematopoietic, and nervous systems; however, it remains unclear whether and how it regulates retinal development. By expression profiling, RNA in situ hybridization and immunostaining, here we show that Ldb1 is expressed in the progenitors during early retinal development, but later its expression gradually shifts to non-photoreceptor cell types including bipolar, amacrine, horizontal, ganglion, and Müller glial cells. Retina-specific ablation of Ldb1 in mice resulted in microphthalmia, optic nerve hypoplasia, retinal thinning and detachment, and profound vision impairment as determined by electroretinography. In the mutant retina, there was precocious differentiation of amacrine and horizontal cells, indicating a requirement of Ldb1 in maintaining the retinal progenitor pool. Additionally, all non-photoreceptor cell types were greatly reduced which appeared to be caused by a generation defect and/or retinal degeneration via excessive cell apoptosis. Furthermore, we showed that misexpressed Ldb1 was sufficient to promote the generation of bipolar, amacrine, horizontal, ganglion, and Müller glial cells at the expense of photoreceptors. Together, these results demonstrate that Ldb1 is not only necessary but also sufficient for the development and/or maintenance of non-photoreceptor cell types, and implicate that the pleiotropic functions of Ldb1 during retinal development are context-dependent and determined by its interaction with diverse LIM-HD (LIM-homeodomain) and LMO (LIM domain-only) binding protein partners.

15.
Mol Neurobiol ; 55(4): 2922-2933, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28456934

RESUMO

Amacrine cells are the most diverse group of retinal neurons. Various subtypes of amacrine interneurons mediate a vast majority of image forming and non-image forming visual functions. The transcriptional regulation governing the development of individual amacrine cell subtypes is not well understood. One such amacrine cell subtype comprises neuronal nitric oxide synthase (nNOS/bNOS/NOS1)-expressing amacrine cells (NOACs) that regulate the release of nitric oxide (NO), a neurotransmitter with physiological and clinical implications in the retina. We have identified the LIM-homeodomain transcription factor LHX9 to be necessary for the genesis of NOACs. During retinal development, NOACs express Lhx9, and Lhx9-null retinas lack NOACs. Lhx9-null retinas also display aberrations in dendritic stratification at the inner plexiform layer. Our cell lineage-tracing studies show that Lhx9-expressing cells give rise to both the GAD65 and GAD67 expressing sub-populations of GABAergic amacrine cells. As development proceeds, Lhx9 is downregulated in the GAD65 sub-population of GABAergic cells and is largely restricted to the GAD67 sub-population of amacrine cells that NOACs are a part of. Taken together, we have uncovered Lhx9 as a new molecular marker that defines a subset of amacrine cells and show that it is necessary for the development of the NOAC subtype of amacrine cells.


Assuntos
Células Amácrinas/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Óxido Nítrico/biossíntese , Retina/citologia , Fatores de Transcrição/metabolismo , Células Amácrinas/citologia , Animais , Linhagem da Célula , Dendritos/metabolismo , Neurônios GABAérgicos/metabolismo , Glutamato Descarboxilase/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo I/metabolismo , Fenótipo , Neurônios Retinianos/metabolismo
16.
Mol Cell Endocrinol ; 434: 250-65, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27402603

RESUMO

Gonadotrope cell identity genes emerge in a stepwise process during mouse pituitary development. Cga, encoding for the α-subunit of TSH, LH, and FSH, is initially detected at E11.5 followed by Gnrhr and steroidogenic factor Sf1 at E13.5, specifying cells engaged in a gonadotrope cell fate. Lhb and Fshb appear at E16.5 and 17.5, respectively, typifying differentiated gonadotrope cells. Using the αT1-1, αT3-1 and LßT2 cell lines recapitulating these stages of gonadotrope differentiation, DNA methylation at Gnrhr and Sf1 was investigated. Regulatory regions were found hypermethylated in progenitor αT1-1 cells and hypomethylated in differentiated LßT2 cells. Abundance of RNA polymerase II together with active histone modifications including H3K4me1, H3K4me3, and H3K27ac were strictly correlated with DNA hypomethylation. Analyses of epigenomic modifications and chromatin accessibility were further extended to Isl1, Lhx3, Gata2, and Pitx2, highlighting alternative usages of specific regulatory gene domains in progenitor αT1-1, immature αT3-1, and mature LßT2 gonadotrope cells.


Assuntos
Metilação de DNA , Elementos Facilitadores Genéticos , Gonadotrofos/citologia , Regiões Promotoras Genéticas , Animais , Diferenciação Celular , Linhagem Celular , Epigênese Genética , Epigenômica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa de Hormônios Glicoproteicos/genética , Gonadotrofos/metabolismo , Camundongos , Fatores de Processamento de RNA/genética , Receptores LHRH/genética
17.
Med Gas Res ; 6(2): 70-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27867470

RESUMO

Postoperative nausea and vomiting (PONV) is a common complication after general anesthesia. Recent studies suggested that the hippocampus is involved in PONV. Hypothesising that hippocampal dopaminergic neurons are related to PONV, we examined the comprehensive mRNA profile of the hippocampus, using a sevoflurane-treated mouse model to confirm this. This study was conducted after approval from our institutional animal ethics committee, the Animal Research Center of Sapporo Medical University School of Medicine (project number: 12-033). Eight mice were assigned to two groups: a naïve group and a sevoflurane group (Sev group). In the Sev group, four mice were anesthetised with 3.5% sevoflurane for 1 hour. Subsequently, mRNA was isolated from their hippocampal cells and RNA sequencing was performed on an Illumina HiSeq 2500 platform. Mapping of the quality-controlled, filtered paired-end reads to mouse genomes and quantification of the expression levels of each gene were performed using R software. The Rtn4rl2 gene that encodes the Nogo receptor was the most up-regulated gene in the present study. The expression levels of dopamine receptor genes and the tachykinin gene were increased by sevoflurane exposure, while the genes related to serotonin receptors were not altered by sevoflurane exposure. The expression levels of LIM-homeodomain-related genes were highly down-regulated by sevoflurane. These findings suggest that sevoflurane exposure induces dopaminergic stimulation of hippocampal neurons and triggers PONV, while neuronal inflammation caused by LIM-homeodomain-related genes is down-regulated by sevoflurane.

18.
Gene Expr Patterns ; 19(1-2): 45-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26220830

RESUMO

Lhx9 is a member of the LIM-homeodomain gene family necessary for the correct development of many organs including gonads, limbs, heart and the nervous system. In the context of limb development, Lhx9 has been implicated as an integrator for Fibroblast growth factor (FGF) and Sonic hedgehog (Shh) signalling required for proximal-distal (PD) and anterior-posterior (AP) development of the limb. Three splice variants of the Lhx9 transcript are expressed during development, two of which are predicted to act in a dominant negative fashion, competing with the DNA binding version of Lhx9 for binding to cofactors via the LIM-domain. We examined the expression pattern for the three alternative splice forms of Lhx9; Lhx9α, Lhx9ß and Lhx9c during early limb development. We have found that of the three Lhx9 isoforms, only Lhx9α and Lhx9c (intact homeodomain) are expressed during early limb development, each with their own distinct expression pattern. Additionally we determined that Lhx9 expression overlaps with FGF10 expression in the developing limb bud mesenchyme. Limb bud explant cultures, in the presence of signalling pathway inhibitors, also indicated that Lhx9 mRNA expression in the limb bud was dependent on FGF signalling.


Assuntos
Fator 10 de Crescimento de Fibroblastos/metabolismo , Proteínas com Homeodomínio LIM/biossíntese , Proteínas com Homeodomínio LIM/genética , Botões de Extremidades/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Animais , Extremidades/embriologia , Feminino , Fator 10 de Crescimento de Fibroblastos/biossíntese , Fator 10 de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/genética , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Botões de Extremidades/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Transativadores/metabolismo
19.
J Biosci Bioeng ; 119(3): 260-6, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25239070

RESUMO

Expression of the LIM homeodomain transcription factor Lhx8 is restricted to and up-regulated in the mesenchyme of the upper face prominence before lip fusion. Msx1/2 acts in early development to control cell proliferation and differentiation. Deficiency of these genes is associated with nonsyndromic cleft lip with/without cleft palate. Since retinoid is a potential patterning influence on the developing face, we have examined whether retinoic acid (RA) signaling regulated Lhx8, Msx1 and Msx2 transcription through fibroblast growth factor (FGF) signals in the maxillary prominence. Application of exogenous RA caused severe defects of the maxilla. Citral also induced a specific loss of derivatives from the maxillary prominences by blocking RA synthesis. Real-time RT-PCR and semi-quantitative RT-PCR analysis of the maxillary mesenchyme revealed that the expressions of Lhx8, Msx1 and Msx2 were significantly down-regulated by RA as well as by citral. The downregulated Lhx8 was rescued by combined treatment with FGF-8b, which indicated a downstream of RA signaling. FGF-8b induced up-regulated Lhx8 expression whereas SU5402, a pan-FGF family antagonist, down-regulated and caused defective maxillary morphogenesis and cleft lip. Our data suggest that Lhx8 is regulated by RA signaling through FGF signals and the level window of RA and FGF-8b could control the upper jaw morphogenesis.


Assuntos
Fator 8 de Crescimento de Fibroblasto/metabolismo , Arcada Osseodentária/efeitos dos fármacos , Arcada Osseodentária/embriologia , Proteínas com Homeodomínio LIM/metabolismo , Fatores de Transcrição/metabolismo , Tretinoína/farmacologia , Monoterpenos Acíclicos , Animais , Diferenciação Celular/efeitos dos fármacos , Embrião de Galinha , Regulação para Baixo/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/antagonistas & inibidores , Fator 8 de Crescimento de Fibroblasto/farmacologia , Proteínas de Homeodomínio/metabolismo , Arcada Osseodentária/metabolismo , Fator de Transcrição MSX1/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Monoterpenos/farmacologia , Morfogênese/efeitos dos fármacos , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos
20.
Dev Growth Differ ; 37(5): 551-557, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37281420

RESUMO

The mec-3 gene, a member of the LIM-homeodomain transcription factors, is required for touch receptor, FLP and PVD neurons to differentiate in the nematode Caenorhabditis elegans. Stably integrated transgenic strains with mec-3-lacZ fusion were generated by irradiating UV light to an unstable transgenic strain with the extrachromosomal DNA. Expression patterns of the mec-3-lacZ fusion were examined in mutant backgrounds (lin-4, lin-14, egl-44, egl-46 and sem-4 genes) which alter touch receptor-specific gene expression. In the lin-4 mutant background, ectopic mec-3-lacZ positive AVM/PVM-like cells were observed in 9% of the animals. By contrast, in the lin-14 mutant background, mec-3-lacZ staining in AVM/PVM cells was lost in 86% of the animals. In the egl-44 and egl-46 mutant backgrounds, expression pattern was the same as wild-type animals. In the sem-4 mutant background, more than half of the animals (54-69%) had ectopic staining cells in the tail in addition to the wild-type staining pattern. The modes of action of these genetically interacting genes in the differentiation of mechanosensory neurons are proposed.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa