Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Cell Mol Med ; 28(7): e18193, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38506205

RESUMEN

Colorectal cancer (CRC) liver metastasis, albeit a stage-IV disease, is completely curable by surgical resection in selected patients. In addressing the molecular basics of this phenomenon, differentially expressed genes at primary and liver metastatic sites were screened by RNA sequencing with the use of paraffin-embedded surgical specimens. Chemokine C-C motif ligand 1 (CCL1), a chemotactic factor for a ligand of the chemokine C-C motif receptor 8 (CCR8), was isolated as one of the differentially expressed genes. Histological analysis revealed that the number of CCL1-positive cells, mainly tumour associated macrophages (TAMs) located in the stroma of CRC, decreased significantly at liver metastatic sites, while the expression level of CCR8 on CRC remained unchanged. To explore the biological significance of the CCL1-CCR8 axis in CRC, CCR8-positive CRC cell line Colo320DM was used to assess the effect of the CCL1-CCR8 axis on major signalling pathways, epithelial mesenchymal transition induction and cell motility. Upon stimulation of recombinant CCL1 (rCCL1), phosphorylation of AKT was observed in Colo320DM cells; on the other hand, the corresponding significant increase in MMP-2 levels demonstrated by RT-qPCR was nullified by siRNA (siCCR8). In the scratch test, rCCL1 treatment significantly increased the motility of Colo320DM cells, which was similarly nullified by siCCR8. Thus, the activation of the CCL1-CCR8 axis is a positive regulator of CRC tumour progression. Reduced CCL1 expression of TAMs at liver metastatic sites may partly explain the unique slow tumour progression of CRC, thus providing for a grace period for radical resection of metastatic lesions.


Asunto(s)
Neoplasias Colorrectales , Hígado , Humanos , Quimiocina CCL1 , Ligandos , Hígado/metabolismo , Quimiocinas , Receptores de Quimiocina/metabolismo , Neoplasias Colorrectales/genética
2.
Histochem Cell Biol ; 159(3): 263-273, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36418613

RESUMEN

Since epigenetic modifications differ from cell to cell, detecting the DNA methylation status of individual cells is requisite. Therefore, it is important to conduct "morphology-based epigenetics research", in which the sequence-specific DNA methylation status is observed while maintaining tissue architecture. Here we demonstrate a novel histochemical technique that efficiently shows the presence of a single methylated cytosine in a sequence-dependent manner by applying ICON (interstrand complexation with osmium for nucleic acids) probes. By optimizing the concentration and duration of potassium osmate treatment, ICON probes selectively hybridize to methylated cytosine on tissue sections. Since the elongation process by rolling-circle amplification through the padlock probe and synchronous amplification by the hyperbranching reaction at a constant temperature efficiently amplifies the reaction, it is possible to specifically detect the presence of a single methylated cytosine. Since the ICON probe is cross-linked to the nuclear or mitochondrial DNA of the target cell, subsequent elongation and multiplication reactions proceed like a tree growing in soil with its roots firmly planted, thus facilitating the demonstration of methylated cytosine in situ. Using this novel ICON-mediated histochemical method, detection of the methylation of DNA in the regulatory region of the RANK gene in cultured cells and of mitochondrial DNA in paraffin sections of mouse cerebellar tissue was achievable. This combined ICON and rolling-circle amplification method is the first that shows evidence of the presence of a single methylated cytosine in a sequence-specific manner in paraffin sections, and is foreseen as applicable to a wide range of epigenetic studies.


Asunto(s)
Citosina , Parafina , Animales , Ratones , Metilación de ADN , Epigénesis Genética , ADN Mitocondrial
3.
Histochem Cell Biol ; 160(5): 407-418, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37532885

RESUMEN

The liver is known to possess remarkable regenerative potential, but persistent inflammation or severe acute injury can lead to liver fibrosis and incomplete regeneration, ultimately resulting in liver failure. Recent studies have shown that the axis of two types of CXCL12 receptors, CXCR4 and CXCR7, plays a crucial role in liver fibrosis and regeneration. The present study aimed to investigate the regulatory factors involved in CXCR4 expression in injured liver. Immunohistochemical screening of liver tissue samples collected during liver transplantation revealed a reciprocal expression pattern between CXCR4 and MeCP2. An in vitro system involving cultured cell lines and H2O2 treatment was established to study the impact of oxidative stress on signaling pathways and epigenetic alterations that affect CXCR4 mRNA expression. Operating through distinct signaling pathways, H2O2 treatment induced a dose-dependent increase in CXCR4 expression in both hepatocyte- and intrahepatic cholangiocyte-derived cells. Treatment of the cells with trichostatin and azacytidine modulated CXCR4 expression in hepatocytes by modifying the methylation status of CpG dinucleotides located in a pair of TA repeats adjacent to the TATA box of the CXCR4 gene promoter. Only MeCP2 bound to oligonucleotides representing the TATA box region when the cytosine residues within the sequence were methylated, as revealed by electrophoretic mobility shift assay (EMSA). Methylation-specific PCR analysis of microdissected samples revealed a correlation between the loss of CpG methylation and the upregulation of CXCR4 in injured hepatocytes, replicating the findings from the in vitro study. Besides the conventional MEK/ERK and NF-κB signaling pathways that activate CXCR4 in intrahepatic cholangiocytes, the unique epigenetic modifications observed in hepatocytes might also contribute to a shift in the CXCR4-CXCR7 balance towards CXCR4, leading to irreversible liver injury and fibrosis. This study highlights the importance of epigenetic modifications in regulating CXCR4 expression in liver injury and fibrosis.


Asunto(s)
Peróxido de Hidrógeno , Receptores CXCR4 , Humanos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática , Regiones Promotoras Genéticas , Desmetilación , Expresión Génica , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/farmacología
4.
Cell Tissue Res ; 388(1): 63-73, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35072792

RESUMEN

Pericytes are pluripotent cells that enclose the endothelium of small blood vessels in the whole body. These cells are thought to play a limited role in vascular development and blood pressure regulation; however, current evidence from numerous studies suggests several significant biologic aspects of pericytes in animals. One viewpoint is that pericytes are also known as potential cellular origin of multiple soft tissue tumors. Experimental evidence of the cellular origin of pericytic tumors is still insufficient, however, and their molecular pathogenesis is poorly understood. Here, we used a conditional constitutively active Smoothened allele (Rosa-SmoM2) and Cre recombinase mice to activate hedgehog (Hh) signaling, exclusively in the monocyte/macrophage and osteoclast lineage (LysMcre) or in RANK expressing cells (RANKcre) that are recognized as osteoclast precursor cells. Mice conditionally expressing SmoM2 with LysMcre displayed no significant skeletal phenotype; surprisingly, however, RANKcre; Rosa-SmoM2 mice frequently developed progressive soft tissue tumors in regions of the leg. Genetic lineage tracing analysis uncovered a new domain of RANKcre-expressing cells in the skeletal muscle interstitial cells that display markers consistent with vascular pericytes. Neoplasms arising from these cells showed increased expression of Matrix metalloproteinases (MMPs) that are molecular indicators of malignancy. Moreover, the tumors displayed strong bone invasive potency associated with osteoclastic bone resorption. Thus, these findings provide a novel insight into tumor pathology: Hh signal activated-pericytes can be a potential cellular origin of multiple soft tissue tumors.


Asunto(s)
Pericitos , Neoplasias de los Tejidos Blandos , Animales , Modelos Animales de Enfermedad , Proteínas Hedgehog/metabolismo , Ratones , Pericitos/metabolismo , Transducción de Señal , Neoplasias de los Tejidos Blandos/patología
5.
Biochem Biophys Res Commun ; 572: 86-91, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34358968

RESUMEN

Receptor Activator of NF-κB (RANK) expressed on osteoclasts and their precursors is a receptor for RANK ligand (RANKL). Signals transduced by RANKL-RANK interaction induce genes essential for the differentiation and function of osteoclasts, partly through the direct binding of NFATc1, to target gene promoters. We have previously cloned a 6-kb fragment containing the 5'-flanking region of the mouse RANK gene and have demonstrated the presence of binding elements of hematological transcription factors, such as MITF, PU.1 and AP-1. Here, we demonstrated the presence of the functional NFATc1 responsive element on the RANK gene promoter. Transfection of an NFATc1-expression vector increased RANK mRNA that was subsequently nullified by NFATc1 knockdown. With the use of electrophoretic mobility shift assay (EMSA), an oligonucleotide (-388/-353) showed specific protein-DNA binding that was blockshifted with an anti-NFATc1 antibody and washed out with excess amounts of the cold consensus sequence. Co-transfection studies with the use of an NFATc1-expression vector and RANK promoter-reporter constructs showed that NFATc1 increased promoter activity 2-fold in RAW264.7 cells that was again nullified as disclosed by mutagenesis studies. Taken together, these results indicate that RANK transcription is positively regulated by the RANKL signal through the direct binding of NFATc1 to its specific binding site of the RANK gene promoter, and suggest the presence of a crucial positive feedback mechanism of gene expression that promotes accelerated terminal differentiation of RANK-positive committed precursors to mature osteoclasts.


Asunto(s)
Factores de Transcripción NFATC/metabolismo , Regiones Promotoras Genéticas/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Células Cultivadas , Retroalimentación Fisiológica , Ratones , Ratones Endogámicos , Factores de Transcripción NFATC/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal/genética
6.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530552

RESUMEN

The reproductive tract in mammals emerges from two ductal systems during embryogenesis: Wolffian ducts (WDs) and Mullerian ducts (MDs). Most of the female reproductive tract (FRT) including the oviducts, uterine horn and cervix, originate from MDs. It is widely accepted that the formation of MDs depends on the preformed WDs within the urogenital primordia. Here, we found that the WD mesenchyme under the regulation of Hedgehog (Hh) signaling is closely related to the developmental processes of the FRT during embryonic and postnatal periods. Deficiency of Sonic hedgehog (Shh), the only Hh ligand expressed exclusively in WDs, prevents the MD mesenchyme from affecting uterine growth along the radial axis. The in vivo cell tracking approach revealed that after WD regression, distinct cells responding to WD-derived Hh signal continue to exist in the developing FRT and gradually contribute to the formation of various tissues such as smooth muscle, endometrial stroma and vascular vessel, in the mouse uterus. Our study thus provides a novel developmental mechanism of FRT relying on WD.


Asunto(s)
Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Proteínas Hedgehog/metabolismo , Organogénesis , Transducción de Señal , Útero/embriología , Útero/metabolismo , Animales , Biomarcadores , Diferenciación Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Modelos Biológicos , Conductos Paramesonéfricos/embriología , Conductos Paramesonéfricos/metabolismo , Organogénesis/genética
7.
Biochem Biophys Res Commun ; 523(4): 961-965, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-31964526

RESUMEN

Low density lipoprotein receptor-related protein 1 (LRP1), a multifunctional cell surface protein, is expressed in bone marrow-derived macrophages. While LRP1 is thought to be a suppressor of osteoclast differentiation at late stages, its function at early stages remains unclear. Here we demonstrate that Lrp1 stable knockdown by lentiviral short hairpin RNA in macrophage cell line RAW264 cells inhibited RANKL-induced osteoclast formation and osteoclastic master transcription factor Nfatc1 mRNA expression as assessed by quantitative RT-PCR. Furthermore, knockdown of the Lrp1 gene suppressed not only differentiation, but also proliferation, and inhibitory effects on osteoblastic ALP activity by osteoclast-derived humoral factors. Thus, we propose that LRP1 in macrophages is required for both differentiation into osteoclasts and osteoclast-osteoblast interactions.


Asunto(s)
Comunicación Celular , Diferenciación Celular , Técnicas de Silenciamiento del Gen , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Osteoblastos/citología , Osteoclastos/citología , Osteoclastos/metabolismo , Animales , Proliferación Celular , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7
8.
Biochem Biophys Res Commun ; 529(2): 186-190, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32703409

RESUMEN

The long bone midshaft expands by forming primary osteons at the periosteal surface of cortical bone in humans and rodents. Osteoblastic bone formation in the vascular cavity in the center of primary osteons is delayed during cortical bone development. The mechanisms of the formation of primary osteons is not fully understood, however. Focusing on NOTCH1 signaling, an inhibitory signaling on osteoblastic bone formation, our immunohistochemical analysis revealed Delta like1 (DLL1), a ligand of NOTCH1, and the NOTCH1 intracellular domain (NICD, an activated form of NOTCH1) immunoreactivity, in the cuboidal osteoblasts lining the bone surface in the vascular cavity of primary osteons during postnatal growth in rats. Interestingly, five days after treatment of primary osteoblasts with ascorbic acid and ß glycerophosphate, protein levels of both DLL1 and NICD increased transiently, indicating that DLL1 activates NOTCH1 in primary cultured osteoblasts. Thus, the results imply that DLL1-NOTCH1 signaling in osteoblasts is associated with primary osteonal bone formation.


Asunto(s)
Hueso Cortical/citología , Péptidos y Proteínas de Señalización Intercelular/análisis , Proteínas de la Membrana/análisis , Osteoblastos/citología , Receptor Notch1/análisis , Animales , Células Cultivadas , Hueso Cortical/metabolismo , Masculino , Osteoblastos/metabolismo , Dominios Proteicos , Ratas , Ratas Wistar
9.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326611

RESUMEN

The functional role of the Hedgehog (Hh)-signaling pathway has been widely investigated in bone physiology/development. Previous studies have, however, focused primarily on Hh functions in bone formation, while its roles in bone resorption have not been fully elucidated. Here, we found that cyclopamine (smoothened (Smo) inhibitor), GANT-58 (GLI1 inhibitor), or GANT-61 (GLI1/2 inhibitor) significantly inhibited RANKL-induced osteoclast differentiation of bone marrow-derived macrophages. Although the inhibitory effects were exerted by cyclopamine or GANT-61 treatment during 0-48 h (early stage of osteoclast differentiation) or 48-96 h (late stage of osteoclast differentiation) after RANKL stimulation, GANT-58 suppressed osteoclast formation only during the early stage. These results suggest that the Smo-GLI1/2 axis mediates the whole process of osteoclastogenesis and that GLI1 activation is requisite only during early cellular events of osteoclastogenesis. Additionally, macrophage/osteoclast-specific deletion of Smo in mice was found to attenuate the aging phenotype characterized by trabecular low bone mass, suggesting that blockage of the Hh-signaling pathway in the osteoclast lineage plays a protective role against age-related bone loss. Our findings reveal a specific role of the Hh-signaling pathway in bone resorption and highlight that its inhibitors show potential as therapeutic agents that block osteoclast formation in the treatment of senile osteoporosis.


Asunto(s)
Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Macrófagos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoporosis/metabolismo , Transducción de Señal/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoporosis/diagnóstico por imagen , Osteoporosis/genética , Piridinas/farmacología , Pirimidinas/farmacología , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptor Smoothened/antagonistas & inhibidores , Receptor Smoothened/metabolismo , Tiofenos/farmacología , Regulación hacia Arriba , Alcaloides de Veratrum/farmacología , Microtomografía por Rayos X , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína Gli2 con Dedos de Zinc/antagonistas & inhibidores , Proteína Gli2 con Dedos de Zinc/metabolismo
10.
Biochem Biophys Res Commun ; 515(2): 268-274, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31146918

RESUMEN

Receptor activator of NF-κB (RANK) expressed on osteoclasts and their precursors is a receptor for RANK ligand (RANKL). Signals transduced by RANKL-RANK interaction induce genes essential for the differentiation and function of osteoclasts. We have cloned a basic promoter region of the mouse RANK gene and have analyzed the transcription machinery by transcription factors such as PU.1 (-480), and MITF (-100). Here, we examined the regulatory mechanisms of RANK gene transcription through AP-1 binding site, agagctca (-240). RANK mRNA expression in pre-osteoclastic RAW264.7 cells was induced by Phorbol12-myristate13-acetate (PMA) and suppressed by protein kinase C (PKC) inhibitor calphostin C. In RAW264.7 cells, Fos knockdown by siRNA blocked the inducible effect of PMA on RANK expression. By EMSA, an oligonucleotide (-246/-238) showed DNA protein binding, the specificity of which was confirmed by block-shift assay with an anti-Fos antibody and by the addition of the excess of a cold consensus probe. Co-transfection with a Fos expression vector showed that Fos increased RANK promoter activity 6-fold in RAW264.7 cells, and the addition of PU.1 and MITF superinduced the activity more than twenty-fold by the addition of PU.1 and MITF. Mutagenesis of the putative AP-1 site (-240) blocked the inducible effect of Fos on promoter activity. Taken together, these results indicate that during the differentiation of bone marrow mono-nucleated cells into osteoclast precursors, RANK transcription is positively regulated by Fos/AP-1 through the binding element of its gene promoter, supporting the concept that Fos activation by continuous CSF-1 stimulation on macrophages triggers initial expression of RANK and, later, a positive feedback loop by RANKL-RANK interaction.


Asunto(s)
Osteogénesis/genética , Osteogénesis/fisiología , Proteína Quinasa C/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Animales , Secuencia de Bases , Sitios de Unión/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Técnicas de Silenciamiento del Gen , Ratones , Mutagénesis Sitio-Dirigida , Osteoclastos/citología , Osteoclastos/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/genética , Ligando RANK/metabolismo , Células RAW 264.7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Elementos de Respuesta , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional
11.
Int J Mol Sci ; 20(23)2019 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-31757091

RESUMEN

The longitudinal growth of long bone, regulated by an epiphyseal cartilaginous component known as the "growth plate", is generated by epiphyseal chondrocytes. The growth plate provides a continuous supply of chondrocytes for endochondral ossification, a sequential bone replacement of cartilaginous tissue, and any failure in this process causes a wide range of skeletal disorders. Therefore, the cellular and molecular characteristics of the growth plate are of interest to many researchers. Hedgehog (Hh), well known as a mitogen and morphogen during development, is one of the best known regulatory signals in the developmental regulation of the growth plate. Numerous animal studies have revealed that signaling through the Hh pathway plays multiple roles in regulating the proliferation, differentiation, and maintenance of growth plate chondrocytes throughout the skeletal growth period. Furthermore, over the past few years, a growing body of evidence has emerged demonstrating that a limited number of growth plate chondrocytes transdifferentiate directly into the full osteogenic and multiple mesenchymal lineages during postnatal bone development and reside in the bone marrow until late adulthood. Current studies with the genetic fate mapping approach have shown that the commitment of growth plate chondrocytes into the skeletal lineage occurs under the influence of epiphyseal chondrocyte-derived Hh signals during endochondral bone formation. Here, we discuss the valuable observations on the role of the Hh signaling pathway in the growth plate based on mouse genetic studies, with some emphasis on recent advances.


Asunto(s)
Desarrollo Óseo , Huesos de la Extremidad Inferior/metabolismo , Huesos de la Extremidad Superior/metabolismo , Placa de Crecimiento/metabolismo , Proteínas Hedgehog/metabolismo , Animales , Huesos de la Extremidad Inferior/crecimiento & desarrollo , Huesos de la Extremidad Superior/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Placa de Crecimiento/crecimiento & desarrollo , Proteínas Hedgehog/genética , Humanos , Transducción de Señal
12.
Int J Mol Sci ; 21(1)2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31861793

RESUMEN

Hedgehog (Hh) signaling is an essential growth factor signaling pathway especially in the regulation of epithelial-mesenchymal interactions (EMI) during the development of the urogenital organs such as the bladder and the external genitalia (EXG). The Hh ligands are often expressed in the epithelia, affecting the surrounding mesenchyme, and thus constituting a form of paracrine signaling. The development of the urogenital organ, therefore, provides an intriguing opportunity to study EMI and its relationship with other pathways, such as hormonal signaling. Cellular interactions of prostate cancer (PCa) with its neighboring tissue is also noteworthy. The local microenvironment, including the bone metastatic site, can release cellular signals which can affect the malignant tumors, and vice versa. Thus, it is necessary to compare possible similarities and divergences in Hh signaling functions and its interaction with other local growth factors, such as BMP (bone morphogenetic protein) between organogenesis and tumorigenesis. Additionally, this review will discuss two pertinent research aspects of Hh signaling: (1) the potential signaling crosstalk between Hh and androgen signaling; and (2) the effect of signaling between the epithelia and the mesenchyme on the status of the basement membrane with extracellular matrix structures located on the epithelial-mesenchymal interface.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas Hedgehog/metabolismo , Neoplasias de la Próstata/metabolismo , Andrógenos/genética , Andrógenos/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Comunicación Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Organogénesis , Neoplasias de la Próstata/genética , Mapas de Interacción de Proteínas , Transducción de Señal , Microambiente Tumoral
13.
Histochem Cell Biol ; 150(1): 3-12, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29721644

RESUMEN

Cytosine methylation plays a major role in the regulation of sequential and tissue-specific expression of genes. De novo aberrant DNA methylation and demethylation are also crucial processes in tumorigenesis and tumor progression. The mechanisms of how and when such aberrant methylation and demethylation occur in tumor cells are still obscure, however. To evaluate subtle epigenetic alteration among minor subclonal populations, morphology-oriented epigenetic analysis is requisite, especially where heterogeneity and flexibility are as notable as in the process of cancer progression and cellular differentiation at critical stages. Therefore, establishment of reliable morphology-oriented epigenetic studies has become increasingly important in not only the experimental but also the diagnostic field. By selecting a subset of cells based on characteristic morphological features disclosed by microdissection or in situ hybridization, we discovered how methylation at certain CpG sites outside of CpG islands would play a crucial epigenetic role in the versatility and flexibility of gene expression during cancer progression. In this review, we first introduce technical aspects of two morphology-oriented epigenetic studies: (1) histoendonuclease-linked detection of methylated sites of DNA (HELMET), and (2) padlock probe and rolling circle amplification (RCA) for in situ identification of methylated cytosine in a sequence-dependent manner. We then present our observation of a novel MeCP2-mediated gene-silencing mechanism through the addition of methylation to a single-CpG-locus upstream of the TATA-box of the receptor activator of NF-κB ligand (RANKL) and of secreted frizzled-related protein 4 (SFRP4) gene promoters.


Asunto(s)
Epigénesis Genética/genética , Neoplasias/genética , Islas de CpG/genética , Citosina/metabolismo , Metilación de ADN , Silenciador del Gen , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo
14.
Histochem Cell Biol ; 149(4): 365-373, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29356962

RESUMEN

Longitudinal bone growth progresses by continuous bone replacement of epiphyseal cartilaginous tissue, known as "growth plate", produced by columnar proliferated- and differentiated-epiphyseal chondrocytes. The endochondral ossification process at the growth plate is governed by paracrine signals secreted from terminally differentiated chondrocytes (hypertrophic chondrocytes), and hedgehog signaling is one of the best known regulatory signaling pathways in this process. Here, to investigate the developmental relationship between longitudinal endochondral bone formation and osteogenic progenitors under the influence of hedgehog signaling at the growth plate, genetic lineage tracing was carried out with the use of Gli1CreERT2 mice line to follow the fate of hedgehog-signal-responsive cells during endochondral bone formation. Gli1CreERT2 genetically labeled cells are detected in hypertrophic chondrocytes and osteo-progenitors at the chondro-osseous junction (COJ); these progeny then commit to the osteogenic lineage in periosteum, trabecular and cortical bone along the developing longitudinal axis. Furthermore, in ageing bone, where longitudinal bone growth ceases, hedgehog-signal responsiveness and its implication in osteogenic lineage commitment is significantly weakened. These results show, for the first time, evidence of the developmental contribution of endochondral progenitors under the influence of epiphyseal chondrocyte-derived secretory signals in longitudinally growing bone. This study provides a precise outline for assessing the skeletal lineage commitment of osteo-progenitors in response to growth-plate-derived regulatory signals during endochondral bone formation.


Asunto(s)
Desarrollo Óseo , Huesos/metabolismo , Placa de Crecimiento/metabolismo , Proteínas Hedgehog/metabolismo , Músculo Esquelético/metabolismo , Transducción de Señal , Animales , Huesos/citología , Placa de Crecimiento/citología , Masculino , Ratones , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
15.
Histochem Cell Biol ; 149(4): 405-415, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29356963

RESUMEN

Hard tissue homeostasis is regulated by the balance between bone formation by osteoblasts and bone resorption by osteoclasts. This physiologic process allows adaptation to mechanical loading and calcium homeostasis. Under pathologic conditions, however, this process is ill-balanced resulting in either over-resorption or over-formation of hard tissue. Local over-resorption by osteoclasts is typically observed in osteolytic metastases of malignancies, autoimmune arthritis, and giant cell tumor of bone (GCTB). In tumor-related local osteolysis, tumor-derived osteoclast-activating factors induce bone resorption not by directly acting on osteoclasts but by indirectly upregulating receptor activator of NFκB ligand (RANKL) on osteoblastic cells. Similarly, synovial tissue in the autoimmune arthritis model does overexpress RANKL and contains numerous osteoclast precursors, and like a landing craft, when it comes in contact with eroded bone surfaces, osteoclast precursors are immediately polarized to become mature osteoclasts, inducing rapidly progressive bone destruction at a late stage of the disease. GCTB, on the other hand, is a common primary bone tumor, usually arising at the metaphysis of the long bone in young adults. After the discovery of RANKL, the concept of GCTB as a tumor of RANKL-expressing stromal cells was established, and comprehensive exosome studies finally disclosed the causative single-point mutation at histone H3.3 (H3F3A) in stromal cells. Thus, osteolytic lesions under various pathological conditions are ultimately attributable to the overexpression of RANKL, which opens up a common, practical and useful therapeutic target for diverse osteolytic conditions.


Asunto(s)
Osteoclastos/patología , Osteólisis/patología , Animales , Humanos , Osteoclastos/metabolismo , Osteólisis/metabolismo , Ligando RANK/metabolismo
16.
Cell Tissue Res ; 359(2): 503-512, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25380565

RESUMEN

Endochondral bone formation is tightly regulated by the spatial and sequential expression of a series of transcription factors. To disclose the roles of TBX18, a member of the T-box transcription factor family, during endochondral bone formation, its spatial and temporal expression patterns were characterized in the limb skeletal region of the developing mouse together with those of established osteochondrogenic markers Sox9, Col2a1, and Runx2. TBX18 expression first appeared in condensed mesenchymal cells (chondro-progenitors) in embryonic-day-10.5 (E10.5) limb bud and was co-localized with Sox9 expression, whereas at E11.5 and E12.5, it became undetectable in mesenchymal cells committed to the chondrocyte lineage. From E13.5 to E18.5, TBX18 expression reappeared in chondrocytes, correlating strongly with Col2a1 expression; furthermore, low level TBX18 expression was found in the Runx2-positive perichondral osteoblastic cell lineage. At the postnatal stage, TBX18 expression was observed in epiphyseal chondrocytes and osteocytes within the lacunae of mature trabecular bone. On the assumption that such characteristic Tbx18 gene expression is epigenetically regulated during mouse limb development, we examined the methylation status of the CpG-island in the mouse Tbx18 gene by methylation-specific polymerase chain reaction. Hypermethylation of the Tbx18 gene promoter became evident at an early embryonic stage in TBX18-negative cells and then disappeared at a late embryonic stage in TBX18-positive cells. Therefore, the temporal suppression of Tbx18 gene expression by the hypermethylation of its promoter seems to trigger the differentiation of mesenchymal cells into hypertrophic chondrocytes in the early stages of endochondral ossification.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Osteogénesis/genética , Proteínas de Dominio T Box/genética , Animales , Secuencia de Bases , Condrogénesis/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Metilación de ADN/genética , Desarrollo Embrionario/genética , Extremidades/embriología , Femenino , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Tiempo
17.
Acta Histochem Cytochem ; 56(3): 41-47, 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37425097

RESUMEN

Keeping chromatin in a stable state is essential for genome stability, scheduled transcription, replication, DNA repair, and precise and reliable chromosome segregation and telomere maintenance during cell division. Over the past decade, research on chromatin remodeling has made great strides whereby modification of histone proteins is a key factor involved in many of the essential cellular processes. The nuclear findings of tumor cells that pathologists routinely examine are nothing but reflections of both genomic and histone alterations. Moreover, impaired histone function is known to be related to common diseases such as diabetes and atherosclerosis, and is, therefore, considered a potential therapeutic target. The present review first outlines the physiological function of histone proteins, and second, demonstrates their alterations to pathological states, emphasizing the importance of immunohistochemistry in histopathological diagnosis.

18.
Development ; 136(23): 3969-78, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19906864

RESUMEN

Embryonic appendicular structures, such as the limb buds and the developing external genitalia, are suitable models with which to analyze the reciprocal interactions of growth factors in the regulation of outgrowth. Although several studies have evaluated the individual functions of different growth factors in appendicular growth, the coordinated function and integration of input from multiple signaling cascades is poorly understood. We demonstrate that a novel signaling cascade governs formation of the embryonic external genitalia [genital tubercle (GT)]. We show that the dosage of Shh signal is tightly associated with subsequent levels of Wnt/beta-catenin activity and the extent of external genitalia outgrowth. In Shh-null mouse embryos, both expression of Wnt ligands and Wnt/beta-catenin signaling activity are downregulated. beta-catenin gain-of-function mutation rescues defective GT outgrowth and Fgf8 expression in Shh-null embryos. These data indicate that Wnt/beta-catenin signaling in the distal urethral epithelium acts downstream of Shh signaling during GT outgrowth. The current data also suggest that Wnt/beta-catenin regulates Fgf8 expression via Lef/Tcf binding sites in a 3' conserved enhancer. Fgf8 induces phosphorylation of Erk1/2 and cell proliferation in the GT mesenchyme in vitro, yet Fgf4/8 compound-mutant phenotypes indicate dispensable functions of Fgf4/8 and the possibility of redundancy among multiple Fgfs in GT development. Our results provide new insights into the integration of growth factor signaling in the appendicular developmental programs that regulate external genitalia development.


Asunto(s)
Genitales/embriología , Proteínas Hedgehog/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis/fisiología , Muerte Celular/fisiología , Línea Celular , Proliferación Celular , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Hedgehog/genética , Inmunohistoquímica , Hibridación in Situ , Integrasas/genética , Integrasas/metabolismo , Luciferasas de Renilla/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Mutantes , Técnicas de Cultivo de Órganos , Plásmidos/genética , Embarazo , Transfección , Proteínas Wnt/genética , beta Catenina/genética
19.
Acta Histochem Cytochem ; 55(1): 1-7, 2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35444348

RESUMEN

Over the past few decades, many researchers have individually identified tumor-related genes, and have accumulated information on their basic research in a database. With the development of technology that can comprehensively test the expression status within a short time, oncogene panel testing has become attainable. On the other hand, changes in gene expression that do not depend on changes in base sequences, that is, epigenetics, or more comprehensively, epigenomes, are also highly involved in the development and progression of disease. Oncogene panel tests tend to focus on DNA base mutations such as point mutations, deletions, duplications, and chimera formation. Elucidation leads to correct interpretation of diseases and treatment choices, and we are in an era where integrated understanding of the genome and epigenome is indispensable. In this review, we make every effort to cover a wide range of knowledge, including data on histone protein modification, non-coding (nc)RNA and DNA methylation, and recent application trials for demonstrating epigenetic alterations in histologic and cytologic specimens. We hope this review will help marshal the knowledge accumulated by researchers involved in genomic and epigenomic studies.

20.
Bone ; 154: 116200, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34534711

RESUMEN

Macrophages are progenitors of osteoclasts as well as regulators of bone metabolism. Macrophages mediate not only bone formation by osteoblasts under physiological conditions, but also bone regeneration after fracture. The mechanisms of macrophages regulation of bone formation and regeneration remain unclear, however. Here, we demonstrate that the liposome-encapsulated Clodronate (Clod-lip) injected mouse model with cortical bone defect induced by drill-hole injury and targeted depletion of phagocytic macrophages exhibits impaired angiogenesis of type H vessels that couple angiogenesis and osteogenesis. Moreover, we identify Tgfbi (encoding TGFBI), Plau (encoding uPA) and Tgfb1 (encoding TGF-ß1), through RNA-seq analysis, as genes of macrophage-secreted factors mediating angiogenesis and wound healing. The relevant mRNA was highly expressed in bone marrow-derived macrophages among bone cells, as determined through qRT-PCR. Finally, we disclose that treatment with uPA inhibitor or TGF-ß receptor I, receptor II inhibitor impairs bone regeneration after injury, confirming the importance of uPA and TGF-ß1 during bone regeneration. Our findings reveal a novel mechanism of bone regeneration mediated by macrophages.


Asunto(s)
Regeneración Ósea , Osteogénesis , Animales , Macrófagos/metabolismo , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda