Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 202(6): 1833-1844, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30700585

RESUMO

The transcription factor Nkx2.3 regulates the vascular specification of Peyer patches in mice through determining endothelial addressin preference and may function as a susceptibility factor in inflammatory bowel diseases in humans. We wished to analyze the role of Nkx2.3 in colonic solitary intestinal lymphoid tissue composition and in colitis pathogenesis. We studied the colonic solitary intestinal lymphoid tissue of Nkx2.3-deficient mice with immunofluorescence and flow cytometry. Colitis was induced in mice using 2.5% dextran sodium sulfate, and severity was assessed with histology, flow cytometry, and quantitative PCR. We found that the lack of Nkx2.3 impairs maturation of isolated lymphoid follicles and attenuates dextran sodium sulfate-induced colitis independent of endothelial absence of mucosal addressin cell-adhesion molecule-1 (MAdCAM-1), which was also coupled with enhanced colonic epithelial regeneration. Although we observed increased numbers of group 3 innate lymphoid cells and Th17 cells and enhanced transcription of IL-22, Ab-mediated neutralization of IL-22 did not abolish the protection from colitis in Nkx2.3-deficient mice. Nkx2.3-/- hematopoietic cells could not rescue wild-type mice from colitis. Using LacZ-Nkx2.3 reporter mice, we found that Nkx2.3 expression was restricted to VAP-1+ myofibroblast-like pericryptal cells. These results hint at a previously unknown stromal role of Nkx2.3 as driver of colitis and indicate that Nkx2.3+ stromal cells play a role in epithelial cell homeostasis.


Assuntos
Colite/imunologia , Proteínas de Homeodomínio/imunologia , Nódulos Linfáticos Agregados/imunologia , Fatores de Transcrição/imunologia , Animais , Colite/metabolismo , Interleucinas/metabolismo , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nódulos Linfáticos Agregados/metabolismo , Células Estromais/imunologia , Fatores de Transcrição/deficiência , Interleucina 22
2.
Cell Rep ; 11(12): 1847-55, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26095368

RESUMO

The ability of hematopoietic stem cells (HSCs) to self-renew is a prerequisite for the establishment of definitive hematopoiesis and life-long blood regeneration. Here, we report the single-stranded DNA-binding transcriptional regulator far upstream element (FUSE)-binding protein 1 (FUBP1) as an essential factor of HSC self-renewal. Functional inactivation of FUBP1 in two different mouse models resulted in embryonic lethal anemia at around E15.5 caused by severely diminished HSCs. Fetal and adult HSCs lacking FUBP1 revealed an HSC-intrinsic defect in their maintenance, expansion, and long-term blood reconstitution, but could differentiate into all hematopoietic lineages. FUBP1-deficient adult HSCs exhibit significant transcriptional changes, including upregulation of the cell-cycle inhibitor p21 and the pro-apoptotic Noxa molecule. These changes caused an increase in generation time and death of HSCs as determined by video-microscopy-based tracking. Our data establish FUBP1 and its recognition of single-stranded genomic DNA as an important element in the transcriptional regulation of HSC self-renewal.


Assuntos
Autorrenovação Celular/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Células-Tronco Fetais/citologia , Células-Tronco Fetais/metabolismo , Regulação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Camundongos , Transdução de Sinais/genética
3.
J Med Genet ; 52(4): 240-7, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25604083

RESUMO

BACKGROUND: SOX9 mutations cause the skeletal malformation syndrome campomelic dysplasia in combination with XY sex reversal. Studies in mice indicate that SOX9 acts as a testis-inducing transcription factor downstream of SRY, triggering Sertoli cell and testis differentiation. An SRY-dependent testis-specific enhancer for Sox9 has been identified only in mice. A previous study has implicated copy number variations (CNVs) of a 78 kb region 517-595 kb upstream of SOX9 in the aetiology of both 46,XY and 46,XX disorders of sex development (DSD). We wanted to better define this region for both disorders. RESULTS: By CNV analysis, we identified SOX9 upstream duplications in three cases of SRY-negative 46,XX DSD, which together with previously reported duplications define a 68 kb region, 516-584 kb upstream of SOX9, designated XXSR (XX sex reversal region). More importantly, we identified heterozygous deletions in four families with SRY-positive 46,XY DSD without skeletal phenotype, which define a 32.5 kb interval 607.1-639.6 kb upstream of SOX9, designated XY sex reversal region (XYSR). To localise the suspected testis-specific enhancer, XYSR subfragments were tested in cell transfection and transgenic experiments. While transgenic experiments remained inconclusive, a 1.9 kb SRY-responsive subfragment drove expression specifically in Sertoli-like cells. CONCLUSIONS: Our results indicate that isolated 46,XY and 46,XX DSD can be assigned to two separate regulatory regions, XYSR and XXSR, far upstream of SOX9. The 1.9 kb SRY-responsive subfragment from the XYSR might constitute the core of the Sertoli-cell enhancer of human SOX9, representing the so far missing link in the genetic cascade of male sex determination.


Assuntos
Variações do Número de Cópias de DNA , Transtornos do Desenvolvimento Sexual/genética , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição SOX9/genética , Animais , Linhagem Celular , Estudos de Coortes , Feminino , Humanos , Masculino , Camundongos , Linhagem
4.
J Immunol ; 193(10): 5284-93, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25320278

RESUMO

Although the homing of lymphocytes to GALT has been extensively studied, little is known about how high endothelial venules (HEVs) within Peyer's patches (PPs) are patterned to display dominantly mucosal addressin cell adhesion molecule 1 (MAdCAM-1). In this study, we report that Nkx2-3-deficient mice show gradual loss of MAdCAM-1 in PPs postnatally and increased levels of mRNA for peripheral lymph node addressin (PNAd) backbone proteins as well as enhanced expression of MECA79 sulfated glycoepitope at the luminal aspect of HEVs, thus replacing MAdCAM-1 with PNAd. Induction of PNAd in mutant PPs requires lymphotoxin ß receptor activity, and its upregulation needs the presence of mature T and B cells. Furthermore, treatment with MECA-79 anti-PNAd mAb in vivo effectively blocks lymphocyte homing to mutant PPs. Despite the replacement of MAdCAM-1 by PNAd in HEV endothelia, lymphocytes could efficiently home to PPs in mutant mice. We conclude that although Nkx2-3 activity controls the addressin balance of HEVs in GALT, the general HEV functionality is preserved independently from Nkx2-3, indicating a substantial plasticity in the specification of GALT HEV endothelium.


Assuntos
Linfócitos B/metabolismo , Proteínas de Homeodomínio/imunologia , Nódulos Linfáticos Agregados/metabolismo , Linfócitos T/metabolismo , Fatores de Transcrição/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais/farmacologia , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/imunologia , Linfonodos/citologia , Linfonodos/imunologia , Linfonodos/metabolismo , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/imunologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucoproteínas , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Vênulas/citologia , Vênulas/imunologia , Vênulas/metabolismo
5.
J Histochem Cytochem ; 59(7): 690-700, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21705651

RESUMO

In contrast to peripheral lymph nodes possessing lymphatic and blood vasculature, the spleen in both humans and rodents is largely devoid of functioning lymphatic capillaries. Here it is reported that in mice lacking homeodomain transcription factor Nkx2-3, the spleen contains an extensive network of lymphocyte-filled sacs lined by cells expressing LYVE-1 antigen, a marker associated with lymphatic endothelium cells (LECs). Real-time quantitative PCR analyses of Nkx2-3 mutant spleen revealed a substantial increase of LYVE-1 and podoplanin mRNA levels, without the parallel increase of mRNA for VEGFR-3 (vascular endothelial growth factor receptor Type 3) and Prox1 (Prospero homeobobox protein 1), two markers specific for LECs. Although these structures express VEGFR-2/flk-1, they lack Prox1 protein, indicating their non-LEC endothelial origin. The LYVE-1(+) structures are bordered with ER-TR7(+) fibroblastic reticular cells with small clusters of macrophages expressing MARCO and sialoadhesin. Short-term cell-tracing studies using labeled lymphocytes indicate that these LYVE-1(+) cysts are largely excluded from the systemic circulation. Cells expressing LYVE-1 glycoprotein as putative precursors for such structures are detectable in the spleen of late-stage embryos, and the formation of LYVE-1(+) structures is independent from the activity of lymphotoxin ß-receptor. Thus the splenic vascular defects in Nkx2-3 deficiency include the generation of LYVE-1(+) cysts, comprised of endothelial cells without being committed along the LEC lineage.


Assuntos
Cistos/patologia , Células Endoteliais/patologia , Endotélio Linfático/patologia , Endotélio Vascular/patologia , Glicoproteínas/metabolismo , Proteínas de Homeodomínio/genética , Baço/irrigação sanguínea , Fatores de Transcrição/genética , Animais , Linhagem da Célula , Cistos/metabolismo , Células Endoteliais/metabolismo , Endotélio Linfático/metabolismo , Endotélio Vascular/metabolismo , Linfócitos/patologia , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/fisiologia , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Transdução de Sinais , Baço/embriologia , Baço/metabolismo
6.
Pathol Oncol Res ; 13(3): 227-35, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17922052

RESUMO

The capacity of secondary lymphoid organs to provide suitable tissue environment for mounting immune responses is dependent on their compartmentalized stromal constituents, including distinct fibroblasts. In addition to various members of the tumor necrosis factor/lymphotoxin beta family as important morphogenic regulators of peripheral lymphoid tissue development, the formation of stromal elements of spleen is also influenced by the Nkx2.3 homeodomain transcription factor in a tissue-specific fashion. Here we extend our previous work on the role of Nkx2.3-mediated regulation in the development of spleen architecture by analyzing the structure of reticular fibroblastic meshwork of spleen in inbred Nkx2.3-deficient mice. Using immunohistochemistry and dual-label immunofluorescence we found both distributional abnormalities, manifested as poor reticular compartmentalization of T-zone and circumferential reticulum, and developmental blockade, resulting in the absence of a complementary fibroblast subpopulation of white pulp. The disregulated distribution of fibroblasts was accompanied with an increased binding of immunohistochemically detectable complement factor C4 by T-cell zone-associated reticular fibroblasts, distinct from follicular dendritic cells with inherently high-level expression of bound C4. These data indicate that the impact of Nkx2.3 gene deficiency on fibroblast ontogeny within the spleen extends beyond its distributional effects, and that the formation of various white pulp fibroblast subsets is differentially affected by the presence of Nkx2.3 activity, possibly also influencing their role in various immune functions linked with complement activation and deposition.


Assuntos
Fibroblastos/patologia , Proteínas de Homeodomínio/genética , Baço/metabolismo , Baço/patologia , Fatores de Transcrição/genética , Animais , Anticorpos Monoclonais/metabolismo , Complemento C4/metabolismo , Células Dendríticas Foliculares/metabolismo , Células Dendríticas Foliculares/patologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Proteínas de Homeodomínio/metabolismo , Sistema Imunitário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fatores de Transcrição/metabolismo
7.
Cell Tissue Res ; 328(3): 473-86, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17318587

RESUMO

The formation of peripheral lymphoid tissues is indispensable for the efficient recognition and elimination of external antigens by lymphoid and accessory cells of the adaptive immune system. The spleen is structurally arranged around various vascular beds with distinct endothelial phenotypes. Using immunohistochemistry, we investigated the postnatal developmental characteristics of the marginal sinus and its relationship with various red-pulp sinus subsets. We also determined the importance of the lymphotoxin beta receptor (LT beta R) and the role of the Nkx2.3 transcription factor for the formation of the splenic vasculature. Both the administration of soluble LT beta R-Ig fusion protein to neonates and the deletion of LT beta R or downstream signaling components (RelB and p52) of the NF-kappaB family inhibited the phenotypic maturation of marginal sinus but had no effect on the vascular compartmentalization of the red pulp. The integrity of the marginal sinus and the proper vascular segregation of the red pulp appeared to be controlled by Nkx2.3, as Nkx2.3-deficient mice exhibited an abnormal distribution of IBL-7/1(hi)/IBL-9/2(-) sinuses and a lack of IBL-7/1(lo)/IBL-9/2(+) vessels. Our data suggest that phenotypic heterogeneity among different vascular elements within distinct anatomical regions of the spleen differentially depends on developmental factors such as lymphotoxin signaling or Nkx2.3, whereas the marginal sinus is controlled by both pathways.


Assuntos
Endotélio Vascular/citologia , Proteínas de Homeodomínio/fisiologia , Linfotoxina-beta/fisiologia , Baço/irrigação sanguínea , Fatores de Transcrição/fisiologia , Animais , Animais Recém-Nascidos , Moléculas de Adesão Celular/metabolismo , Diferenciação Celular/genética , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Receptor beta de Linfotoxina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos SCID , Modelos Biológicos , Mucoproteínas , Transdução de Sinais/fisiologia , Baço/citologia , Baço/metabolismo , Fatores de Transcrição/genética
8.
FEBS Lett ; 579(20): 4254-8, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16051233

RESUMO

Raver2 was identified as a novel member of the hnRNP family based on sequence homology within three RNA recognition motifs and its general domain organization reminiscent of the previously described raver1 protein. Like raver1, raver2 contains two putative nuclear localization signals and a potential nuclear export sequence, and also displays nucleo-cytoplasmic shuttling in a heterokaryon assay. In glia cells and neurons, raver2 localizes to the nucleus. Moreover, the protein interacts with polypyrimidine tract binding protein (PTB) suggesting that it may participate in PTB-mediated nuclear functions. In contrast to ubiquitously expressed raver1, raver2 exerts a distinct spatio-temporal expression pattern during embryogenesis and is essentially restricted to brain, lung, and kidney in the adult mouse.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/química , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/metabolismo , Núcleo Celular/química , Núcleo Celular/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Ribonucleoproteínas Nucleares Heterogêneas/classificação , Humanos , Camundongos , Dados de Sequência Molecular , Neuroglia/citologia , Neurônios/química , Neurônios/metabolismo , Sinais de Localização Nuclear , Proteínas Nucleares/metabolismo , Proteína de Ligação a Regiões Ricas em Polipirimidinas/análise , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Transporte Proteico , Proteínas de Ligação a RNA , Ribonucleoproteínas , Distribuição Tecidual
9.
J Biol Chem ; 278(48): 48377-85, 2003 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-13129914

RESUMO

Lis1 protein is the non-catalytic component of platelet-activating factor acetylhydrolase 1b (PAF-AH 1B) and associated with microtubular structures. Hemizygous mutations of the LIS1 gene cause type I lissencephaly, a brain abnormality with developmental defects of neuronal migration. Lis1 is also expressed in testis, but its function there has not been determined. We have generated a mouse mutant (LIS1GT/GT) by gene trap integration leading to selective disruption of a Lis1 splicing variant in testis. Homozygous mutant males are infertile with no other apparent phenotype. We demonstrate that Lis1 is predominantly expressed in spermatids, and spermiogenesis is blocked when Lis1 is absent. Mutant spermatids fail to form correct acrosomes and nuclei appear distorted in size and shape. The tissue architecture in mutant testis appears severely disturbed displaying collapsed seminiferous tubules, mislocated germ cells, and increased apoptosis. These results provide evidence for an essential and hitherto uncharacterized role of the Lis1 protein in spermatogenesis, particularly in the differentiation of spermatids into spermatozoa.


Assuntos
Infertilidade Masculina/etiologia , Proteínas Associadas aos Microtúbulos/biossíntese , Espermátides/metabolismo , Testículo/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase , Acrossomo/metabolismo , Animais , Apoptose , Northern Blotting , Western Blotting , Diferenciação Celular , Núcleo Celular/metabolismo , Fragmentação do DNA , Modelos Animais de Doenças , Dineínas/biossíntese , Éxons , Feminino , Biblioteca Gênica , Genótipo , Homozigoto , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Modelos Genéticos , Mutação , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatogênese , Tubulina (Proteína)/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA