Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Genesis ; 61(1-2): e23509, 2023 03.
Article in English | MEDLINE | ID: mdl-36622051

ABSTRACT

Craniofacial anomalies (CFAs) are a diverse group of disorders affecting the shapes of the face and the head. Malformation of the cranial base in humans leads CFAs, such as midfacial hypoplasia and craniosynostosis. These patients have significant burdens associated with breathing, speaking, and chewing. Invasive surgical intervention is the current primary option to correct these structural deficiencies. Understanding molecular cellular mechanism for craniofacial development would provide novel therapeutic options for CFAs. In this study, we found that enhanced bone morphogenetic protein (BMP) signaling in cranial neural crest cells (NCCs) (P0-Cre;caBmpr1a mice) causes premature fusion of intersphenoid synchondrosis (ISS) resulting in leading to short snouts and hypertelorism. Histological analyses revealed reduction of proliferation and higher cell death in ISS at postnatal day 3. We demonstrated to prevent the premature fusion of ISS in P0-Cre;caBmpr1a mice by injecting a p53 inhibitor Pifithrin-α to the pregnant mother from E15.5 to E18.5, resulting in rescue from short snouts and hypertelorism. We further demonstrated to prevent premature fusion of cranial sutures in P0-Cre;caBmpr1a mice by injecting Pifithrin-α through E8.5 to E18.5. These results suggested that enhanced BMP-p53-induced cell death in cranial NCCs causes premature fusion of ISS and sutures in time-dependent manner.


Subject(s)
Craniofacial Abnormalities , Skull Base , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neural Crest/pathology , Cell Proliferation , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Male , Female , Animals , Mice , Animals, Newborn , Signal Transduction , Apoptosis , Chondrocytes/metabolism , Smad Proteins/metabolism , Protein Binding , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Pregnancy , Skull Base/abnormalities , Skull Base/metabolism , Skull Base/pathology , Hypertelorism/metabolism , Hypertelorism/pathology
2.
Genesis ; 59(5-6): e23419, 2021 06.
Article in English | MEDLINE | ID: mdl-33851764

ABSTRACT

BMP signaling plays pleiotropic roles in various tissues during embryogenesis and after birth. We have previously generated a constitutively activated Acvr1(ca-Acvr1) transgenic mouse line (line L35) through pronuclei injection to investigate impacts of enhanced BMP signaling in a tissue specific manner. However, line L35 shows a restricted expression pattern of the transgene. Here, we generated another ca-Acvr1 transgenic line, line A11, using embryonic stem (ES) transgenesis. The generated line A11 shows distinctive phenotypes from line L35, along with very limited expression levels of the transgene. When the transgene is activated in the neural crest cells in a Cre-dependent manner, line A11 exhibits cleft palate and shorter jaws, while line L35 develops ectopic cartilages and highly hypomorphic facial structures. When activated in limb buds, line A11 develops organized but smaller limb skeletal structures, while line L35 forms disorganized limbs with little mineralization. Additionally, no heterotopic ossification (HO) is identified in line A11 when bred with NFATc1-Cre mice even after induction of tissue injury, which is an established protocol for HO for line L35. Therefore, the newly generated conditional ca-Acvr1 mouse line A11 provides an additional resource to dissect highly context dependent functions of BMP signaling in development and disease.


Subject(s)
Activin Receptors, Type I/genetics , Bone Morphogenetic Proteins/metabolism , Gene Targeting/methods , Genetic Pleiotropy , Transgenes , Activin Receptors, Type I/metabolism , Animals , Cartilage/metabolism , Chondrogenesis , Limb Buds/embryology , Limb Buds/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction , Transcriptional Activation
3.
Genesis ; 59(10): e23450, 2021 10.
Article in English | MEDLINE | ID: mdl-34487426

ABSTRACT

Podoplanin, PDPN, is a mucin-type transmembrane glycoprotein widely expressed in many tissues, including lung, kidney, lymph nodes, and mineralized tissues. Its function is critical for lymphatic formation, differentiation of type I alveolar epithelial lung cells, and for bone response to biomechanical loading. It has previously been shown that Pdpn null mice die at birth due to respiratory failure emphasizing the importance of Pdpn in alveolar lung development. During the course of generation of Pdpn mutant mice, we found that most Pdpn null mice in the 129S6 and C57BL6/J mixed genetic background die at the perinatal stage, similar to previously published studies with Pdpn null mice, while all Pdpn null mice bred with Swiss outbred mice survived. Surviving mutant mice in the 129S6 and C57BL6/J mixed genetic background showed alterations in the osteocyte lacunocanalicular network, especially reduced osteocyte canaliculi in the tibial cortex with increased tibial trabecular bone. However, adult Pdpn null mice in the Swiss outbred background showed no overt differences in their osteocyte lacunocnalicular network, bone density, and no overt differences when challenged with exercise. Together, these data suggest that genetic variations present in the Swiss outbred mice compensate for the loss of function of PDPN in lung, kidney, and bone.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cell Differentiation/genetics , Lymphangiogenesis/genetics , Membrane Glycoproteins/genetics , Animals , Calcification, Physiologic/genetics , Cancellous Bone/growth & development , Cancellous Bone/metabolism , Gene Expression Regulation, Developmental/genetics , Kidney/growth & development , Lung/growth & development , Lung/metabolism , Lymph Nodes/growth & development , Mice , Osteocytes/metabolism , Tibia/growth & development , Tibia/metabolism
4.
Zoolog Sci ; 36(2): 105-111, 2019 04 01.
Article in English | MEDLINE | ID: mdl-31120644

ABSTRACT

Mammalian T cell death-associated gene 8 (TDAG8)s are activated by extracellular protons. In the present study, we examined whether the TDAG8 homologs of other species are activated by protons as they are in mammals. We found that Xenopus TDAG8 also stimulated cAMP response element (CRE)-driven promoter activities reflecting the activation of Gs/cAMP signaling pathways when they are stimulated by protons. On the other hand, the activities of chicken and zebrafish TDAG8s are hardly affected by protons. Results using chimeric receptors of human and zebrafish TDAG8s indicate that the specificity of the proton-induced activation lies in the extracellular region. These results suggest that protons are not an evolutionarily conserved agonist of TDAG8.


Subject(s)
Protons , Receptors, G-Protein-Coupled/genetics , Animals , Chickens , Cyclic AMP/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Receptors, G-Protein-Coupled/metabolism , Xenopus , Zebrafish
5.
Cell Tissue Res ; 372(1): 161-162, 2018 04.
Article in English | MEDLINE | ID: mdl-29480457

ABSTRACT

The published online version contains mistake in Table 1, Table 2, and some data in Materials and Methods.

6.
Cell Tissue Res ; 372(1): 77-90, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29130118

ABSTRACT

In the pituitary gland, S100ß-positive cells localize in the neurohypophysis and adenohypophysis but the lineage of the two groups remains obscure. S100ß is often observed in many neural crest-derived cell types. Therefore, in this study, we investigate the origin of pituitary S100ß-positive cells by immunohistochemistry for SOX10, a potent neural crest cell marker, using S100ß-green fluorescence protein-transgenic rats. On embryonic day 21.5, a SOX10-positive cell population, which was also positive for the stem/progenitor cell marker SOX2, emerged in the pituitary stalk and posterior lobe and subsequently expanded to create a rostral-caudal gradient on postnatal day 3 (P3). Thereafter, SOX10-positive cells appeared in the intermediate lobe by P15, localizing to the boundary facing the posterior lobe, the gap between the lobule structures and the marginal cell layer, a pituitary stem/progenitor cell niche. Subsequently, there was an increase in SOX10/S100ß double-positive cells; some of these cells in the gap between the lobule structures showed extended cytoplasm containing F-actin, indicating a feature of migration activity. The proportion of SOX10-positive cells in the postnatal anterior lobe was lower than 0.025% but about half of them co-localized with the pituitary-specific progenitor cell marker PROP1. Collectively, the present study identified that one of the lineages of S100ß-positive cells is a SOX10-positive one and that SOX10-positive cells express pituitary stem/progenitor cell marker genes.


Subject(s)
Embryonic Development , Pituitary Gland/embryology , Pituitary Gland/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , SOXE Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Movement , Cell Proliferation , Embryo, Mammalian/metabolism , Green Fluorescent Proteins/metabolism , Pituitary Gland/cytology , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Rats, Wistar , SOXB1 Transcription Factors/metabolism
7.
Cell Tissue Res ; 369(2): 353-368, 2017 08.
Article in English | MEDLINE | ID: mdl-28364143

ABSTRACT

The adenohypophysis comprises six types of endocrine cells, including PIT1-lineage cells such as growth hormone (GH)-producing cells and heterogeneous non-endocrine cells, such as pituitary stem/progenitor cells as a source of endocrine cells. We determine the expression of characteristic stem cell marker genes, including sex-determining region Y-box 2 (Sox2), in mouse pituitary-derived non-endocrine cell lines Tpit/E, Tpit/F1 and TtT/GF. We observed high expression of fibroblast growth factor (FGF) receptors in Tpit/F1 cells, which we characterised by cultivation in medium containing a basic FGF and B27 supplement as used for neural stem-cell differentiation. A 4-day cultivation of Tpit/F1 produced floating embryonic stem-cell-like clumps accompanied by a three-fold increase in Sox2 expression. Passages in these clumps maintained the proliferative activity and Sox2 expression levels. After 10 days of cultivation, Tpit/F1 cell clumps were immuno-positive for SOX2 and Ki67 (proliferation marker) and loosely attached to the well bottom. An additional 10 days of cultivation induced the emergence of GH-positive/pituitary-specific transcription factor (PIT1)-negative cells showing migration from the clumps. Pit1 overexpression in attached cells could not induce GH production. Finally, we confirmed the presence of PIT1-negative GH-producing cells (3.2-7.7 % of all GH-positive cells) in rat pituitary. Thus, we demonstrate that Tpit/F1 has the plasticity to differentiate into one type of hormone-producing cell.


Subject(s)
Cell Differentiation , Endocrine Cells/cytology , Growth Hormone/biosynthesis , Pituitary Gland/cytology , Animals , Biomarkers/metabolism , Cell Adhesion , Cell Aggregation , Cell Line , Cell Movement , Cell Proliferation , Cell Shape , Chromogranins/metabolism , Culture Media , Endocrine Cells/metabolism , Gene Expression Regulation , Mice , Rats , Receptors, Fibroblast Growth Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism
8.
J Anat ; 230(3): 373-380, 2017 03.
Article in English | MEDLINE | ID: mdl-28026856

ABSTRACT

The anterior pituitary originates from the adenohypophyseal placode. Both the preplacode region and neural crest (NC) derive from subdivision of the neural border region, and further individualization of the placode domain is established by a reciprocal interaction between placodal precursors and NC cells (NCCs). It has long been known that NCCs are present in the adenohypophysis as interstitial cells. A recent report demonstrated that NCCs also contribute to the formation of pericytes in the developing pituitary. Here, we attempt to further clarify the role of NCCs in pituitary development using P0-Cre/EGFP reporter mice. Spatiotemporal analyses revealed that GFP-positive NCCs invaded the adenohypophysis in a stepwise manner. The first wave was detected on mouse embryonic day 9.5 (E9.5), when the pituitary primordium begins to be formed by adenohypophyseal placode cells; the second wave occurred on E14.5, when vasculogenesis proceeds from Atwell's recess. Finally, fate tracing of NCCs demonstrated that NC-derived cells in the adenohypophysis terminally differentiate into all hormone-producing cell lineages as well as pericytes. Our data suggest that NCCs contribute to pituitary organogenesis and vasculogenesis in conjunction with placode-derived pituitary stem/progenitor cells.


Subject(s)
Neural Crest/growth & development , Organogenesis/physiology , Pituitary Gland/embryology , Animals , Embryo, Mammalian , Immunohistochemistry , Mice , Mice, Transgenic , Models, Animal
9.
J Reprod Dev ; 63(1): 37-44, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-27773885

ABSTRACT

PROP1 is a pituitary specific transcription factor that plays a crucial role in pituitary organogenesis. The Prop1 shows varied expression patterns that promptly emerge and then fade during the early embryonic period. However, the regulatory mechanisms governing Prop1 expression remain unclear. Here, we investigated whether Prop1 was under epigenetic regulation by DNA methylation. Bisulfite sequencing was performed on DNA obtained from the pituitary glands and livers of rats on embryonic days (E) 13.5 and E14.5, and postnatal days (P) 4 and P30. The methylation of CpG sites in seven regions from 3-kb upstream of the Prop1 transcription start site through to its second intron were examined. Certain differences in CpG-methylation levels were observed in Region-1 (-2772 b to -2355 b), Region-4 (-198 b to +286 b), Region-5 (+671 b to +990 b), and Region-6 (+1113 b to +1273 b) based on comparisons between pituitary and liver DNA on E13.5. DNA methylation in pituitary glands on E14.5, P4, and P30 was generally similar to that observed in in the pituitary gland on E13.5, whereas the anterior and intermediate lobes of the pituitary gland on P4 and P30 showed only small differences. These results indicate that Prop1 is under regulation by CpG methylation during the early period of pituitary primordium development around E13.5.


Subject(s)
DNA Methylation , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Pituitary Gland/embryology , Pituitary Gland/physiology , Animals , Computational Biology , CpG Islands , DNA/metabolism , Epigenesis, Genetic , Female , Gene Expression Profiling , Liver/metabolism , Male , Rats , Rats, Wistar , Temperature
10.
Cell Tissue Res ; 364(2): 273-88, 2016 05.
Article in English | MEDLINE | ID: mdl-26613603

ABSTRACT

The pituitary gland, an indispensable endocrine organ that synthesizes and secretes pituitary hormones, develops with the support of many factors. Among them, neuronatin (NNAT), which was discovered in the neonatal mouse brain as a factor involved in neural development, has subsequently been revealed to be coded by an abundantly expressing gene in the pituitary gland but its role remains elusive. We analyze the expression profile of Nnat and the localization of its product during rat pituitary development. The level of Nnat expression was high during the embryonic period but remarkably decreased after birth. Immunohistochemistry demonstrated that NNAT appeared in the SOX2-positive stem/progenitor cells in the developing pituitary primordium on rat embryonic day 11.5 (E11.5) and later in the majority of SOX2/PROP1 double-positive cells on E13.5. Thereafter, during pituitary embryonic development, Nnat expression was observed in some stem/progenitor cells, proliferating cells and terminally differentiating cells. In postnatal pituitaries, NNAT-positive cells decreased in number, with most coexpressing Sox2 or Pit1, suggesting a similar role for NNAT to that during the embryonic period. NNAT was widely localized in mitochondria, peroxisomes and lysosomes, in addition to the endoplasmic reticulum but not in the Golgi. The present study thus demonstrated the variability in expression of NNAT-positive cells in rat embryonic and postnatal pituitaries and the intracellular localization of NNAT. Further investigations to obtain functional evidence for NNAT are a prerequisite.


Subject(s)
Gene Expression Regulation, Developmental , Membrane Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Pituitary Gland/embryology , Pituitary Gland/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Line , Endoplasmic Reticulum/metabolism , Homeodomain Proteins/metabolism , Lysosomes/metabolism , Male , Mitochondria/metabolism , Peroxisomes/metabolism , Rats , Rats, Wistar , SOXB1 Transcription Factors/metabolism
11.
J Reprod Dev ; 62(3): 241-8, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-26853788

ABSTRACT

The LIM-homeobox transcription factors LHX2 and LHX3s (LHX3a and LHX3b) are thought to be involved in regulating the pituitary glycoprotein hormone subunit genes Cga and Fshß. These two factors show considerable differences in their amino acid sequences for DNA binding and protein-protein interactions and in their vital function in pituitary development. Hence, we compared the DNA binding properties and transcriptional activities of Cga and Fshß between LHX2 and LHX3s. A gel mobility shift assay for approximately 1.1 kb upstream of Cga and 2.0 kb upstream of Fshß varied in binding profiles between LHX2 and LHX3s. DNase I footprinting revealed DNA binding sites in 8 regions of the Cga promoter for LHX2 and LHX3s with small differences in the binding range and strength. In the Fshß promoter, 14 binding sites were identified for LHX2 and LHX3, respectively. There were alternative binding sites to either gene in addition to similar differences observed in the Cga promoter. The transcriptional activities of LHX2 and LHX3s according to a reporter assay showed cell-type dependent activity with repression in the pituitary gonadotrope lineage LßT2 cells and stimulation in Chinese hamster ovary lineage CHO cells. Reactivity of LHX2 and LHX3s was observed in all regions, and differences were observed in the 5'-upstream region of Fshß. However, immunohistochemistry showed that LHX2 resides in a small number of gonadotropes in contrast to LHX3. Thus, LHX3 mainly controls Cga and Fshß expression.


Subject(s)
Follicle Stimulating Hormone, beta Subunit/metabolism , Glycoprotein Hormones, alpha Subunit/metabolism , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , CHO Cells , Cell Line , Cricetinae , Cricetulus , Deoxyribonuclease I/metabolism , Follicle Stimulating Hormone, beta Subunit/genetics , Glycoprotein Hormones, alpha Subunit/genetics , Immunohistochemistry , Mice , Pituitary Gland/metabolism , Promoter Regions, Genetic , Protein Domains , Swine
12.
J Reprod Dev ; 62(1): 93-102, 2016.
Article in English | MEDLINE | ID: mdl-26640231

ABSTRACT

Pituitary-specific transcription factor PROP1, a factor important for pituitary organogenesis, appears on rat embryonic day 11.5 (E11.5) in SOX2-expressing stem/progenitor cells and always coexists with SOX2 throughout life. PROP1-positive cells at one point occupy all cells in Rathke's pouch, followed by a rapid decrease in their number. Their regulatory factors, except for RBP-J, have not yet been clarified. This study aimed to use the 3 kb upstream region and 1st intron of mouse prop1 to pinpoint a group of factors selected on the basis of expression in the early pituitary gland for expression of Prop1. Reporter assays for SOX2 and RBP-J showed that the stem/progenitor marker SOX2 has cell type-dependent inhibitory and activating functions through the proximal and distal upstream regions of Prop1, respectively, while RBP-J had small regulatory activity in some cell lines. Reporter assays for another 39 factors using the 3 kb upstream regions in CHO cells ultimately revealed that 8 factors, MSX2, PAX6, PIT1, PITX1, PITX2, RPF1, SOX8 and SOX11, but not RBP-J, regulate Prop1 expression. Furthermore, a synergy effect with SOX2 was observed for an additional 10 factors, FOXJ1, HES1, HEY1, HEY2, KLF6, MSX1, RUNX1, TEAD2, YBX2 and ZFP36Ll, which did not show substantial independent action. Thus, we demonstrated 19 candidates, including SOX2, to be regulatory factors of Prop1 expression.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Genes, Reporter , Genetic Vectors , Immunohistochemistry , In Situ Hybridization , Introns , Mice , Organogenesis , Pituitary Gland/metabolism , Rats , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology
13.
Cell Tissue Res ; 361(2): 557-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25795141

ABSTRACT

We have recently shown that cells positive for the paired-related homeobox transcription factors PRRX1 and PRRX2 occur in the rat pituitary, and that they are derived from two different origins: pituitary-derived cells positive for stem cell marker SOX2 and extra-pituitary-derived cells negative for SOX2. In this study, we have further characterized the PRRX1- and PRRX2-positive cells that originate from extra-pituitary cells. Immunohistochemical analyses were performed with specific antibodies against PRRX1 and PRRX2 in order to clarify their roles in pituitary vasculogenesis. PRRX1- and PRRX2-positive cells were found in Atwell's recess and at the periphery of the pituitary on embryonic day 15.5 (E15.5). Several PRRX1-positive cells then invaded the anterior lobe, together with a few PRRX2-positive cells, on E16.5. Some PRRX1-positive cells were also positive for mesenchymal stem cell marker NESTIN. Moreover, some PRRX1/NESTIN double-positive cells showed characteristics of vascular endothelial cells with an Isolectin-B4-binding capacity. PRRX1 co-localized with vascular smooth muscle cell/pericyte marker α-smooth muscle actin in the deep area of Atwell's recess. We confirmed the presence of PRRX2/NESTIN double-positive cells at an entry area in Atwell's recess and at the periphery of the pituitary, but PRRX2 did not co-localize with Isolectin B4 or α-smooth muscle actin. These data suggest that PRRX1- and PRRX2-positive mesenchymal stem/progenitor cells are present at the periphery of the embryonic pituitary and at the entry from Atwell's recess and participate in pituitary vasculogenesis by differentiation into vascular endothelial cells and pericytes, whereas the presence of PRRX2 indicates much higher stemness than PRRX1.


Subject(s)
Homeodomain Proteins/analysis , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Pituitary Gland/embryology , Rats/embryology , Animals , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Homeodomain Proteins/metabolism , Pericytes/cytology , Pericytes/metabolism , Pituitary Gland/blood supply , Pituitary Gland/cytology , Rats, Wistar , SOXB1 Transcription Factors/analysis , SOXB1 Transcription Factors/metabolism , Transcription Factors
14.
Cell Tissue Res ; 359(3): 755-66, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25480420

ABSTRACT

We have recently reported that Sox2-expressing pituitary stem/progenitor cells contact each other via a tight-junction protein CAR to form stem/progenitor cell niches in the marginal cell layer facing the lumen and in the clusters scattered in the parenchyma of the anterior lobe. However, the microenvironment of the niche for the maintenance of stem cell function in the pituitary remains obscure. In this study of pituitary stem/progenitor cell niches, we have attempted to identify the expression of juxtacrine factor ephrin and its receptor. We have found that ephrin-B2 is expressed in the pituitary throughout development but changes its localization pattern. Notably, in the adult pituitary, ephrin-B2 immuno-signals occur in SOX2-, E-cadherin-, and CAR-triple-positive stem/progenitor cells in the niches. Our data suggest that ephrin-B2 signaling has an important role in the formation of pituitary stem/progenitor cell niches and in pituitary organogenesis.


Subject(s)
Ephrin-B2/metabolism , Pituitary Gland/cytology , Stem Cell Niche , Stem Cells/metabolism , Aging/metabolism , Animals , Biomarkers/metabolism , Cadherins/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hormones/metabolism , Male , Pituitary Gland/embryology , Pituitary Gland/growth & development , Protein Transport , Rats , S100 Calcium Binding Protein beta Subunit/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology
15.
Cell Tissue Res ; 357(1): 323-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24770895

ABSTRACT

Paired-related homeobox transcription factors, PRRX1 and PRRX2, are known to be important factors for craniofacial and limb morphogenesis. We recently cloned Prrx2 from the porcine adult pituitary cDNA library and found that only PRRX1 is present in the rat embryonic pituitary. In this study, we re-investigated the temporospatial expression and localization of PRRX1 and PRRX2 in the rat pituitary throughout life. The persistent expression of Prrx1 was ascertained after the middle stage of embryonic development, whereas significant expression of Prrx2 was found only in the postnatal pituitary. Immunohistochemistry confirmed that PRRX1-positive cells appeared inside the pituitary on embryonic day 16.5 in the marginal cell layer (MCL), a pituitary stem/progenitor cell niche, and the expanding parenchyma of the anterior pituitary. In contrast, PRRX2-positive cells first appeared in the anterior lobe and intermediate lobe sides of the MCL around postnatal day 30 when the postnatal pituitary growth wave had almost terminated. Immunostaining for PRRX1 with a stem/progenitor cell marker SOX2, a pituitary progenitor marker PROP1, or pituitary hormones revealed that PRRX1 localized in cells in the transition process from the multipotent progenitor stage to the early stage of terminal differentiation throughout life. PRRX2 emerged in cells positive for SOX2 but negative for PROP1 in the anterior and intermediate lobe sides of the postnatal MCL. Thus, PRRX1 and PRRX2 might participate distinctly in pituitary organogenesis and the postnatal cell-supply system.


Subject(s)
Homeodomain Proteins/physiology , Organogenesis/physiology , Pituitary Gland/growth & development , Animals , Cell Differentiation , Cell Proliferation/physiology , Female , Male , Pituitary Gland/cytology , Pituitary Gland/metabolism , Pregnancy , Rats , Rats, Wistar
16.
Cell Tissue Res ; 357(3): 757-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24770897

ABSTRACT

Chemokines are mostly small secreted polypeptides whose signals are mediated by seven trans-membrane G-protein-coupled receptors. Their functions include the control of leukocytes and the intercellular mediation of cell migration, proliferation, and adhesion in several tissues. We have previously revealed that the CXC chemokine ligand 12 (CXCL12) and its receptor 4 (CXCR4) are expressed in the anterior pituitary gland, and that the CXCL12/CXCR4 axis evokes the migration and interconnection of S100ß-protein-positive cells (S100ß-positive cells), which do not produce classical anterior pituitary hormones. However, little is known of the cells producing the other CXCLs and CXCRs or of their characteristics in the anterior pituitary. We therefore examined whether CXCLs and CXCRs occurred in the rat anterior pituitary lobe. We used reverse transcription plus the polymerase chain reaction to analyze the expression of Cxcl and Cxcr and identified the cells that expressed Cxcl by in situ hybridization. Transcripts of Cxcl10 and its receptor (Cxcr3 and toll-like receptor 4, Tlr4) were clearly detected: cells expressing Cxcl10 and Tlr4 were identified amongst S100ß-positive cells and those expressing Cxcr3 amongst adrenocorticotropic hormone (ACTH)-producing cells. We also investigated Cxcl10 expression in subpopulations of S100ß-positive cells. We separated cultured S100ß-positive cells into the round-type (dendritic-cell-like) and process-type (astrocyte- or epithelial-cell-like) by their adherent activity to laminin, a component of the extracellular matrix; CXCL10 was expressed only in round-type S100ß-positive cells. Thus, CXCL10 produced by a subpopulation of S100ß-positive cells probably exerts an autocrine/paracrine effect on S100ß-positive cells and ACTH-producing cells in the anterior lobe.


Subject(s)
Chemokine CXCL10/metabolism , Dendritic Cells/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Animals , Cells, Cultured , Chemokine CXCL12/metabolism , Ligands , Male , Protein Transport , Rats, Transgenic , Rats, Wistar , Receptors, Chemokine/metabolism
17.
Cell Tissue Res ; 357(3): 767-79, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24842050

ABSTRACT

Some non-endocrine cells in the pituitary anterior lobe are responsible for providing stem/progenitor cells to maintain hormone-producing cells. In particular, cells expressing S100ß protein, a calcium-binding protein, have been hypothesized to be a pituitary cell resource. Accumulating data have revealed that S100ß-positive cells comprise heterogeneous populations and some of them certainly show stem/progenitor characteristics in vivo. Hence, we examine whether S100ß-positive cells have the capacity to differentiate into endocrine cells, by means of in vivo and in vitro experiments on transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of the S100ß promoter. Immunohistochemistry of the pituitary confirmed that some S100ß-positive cells expressed SOX2 (SRY [sex-determining region Y]-box 2) and had proliferative activity. Dispersed anterior lobe cells were observed by time-lapse microscopy, followed by immunostaining for hormone and pituitary-transcription-factor1 (PIT1). First, the dispersed anterior lobe cells were immunostained by an antibody against SOX2. S100ß-protein co-localizes with SOX2 (about 89 %). Although 44 of 134 S100ß-positive cells traced were proliferative but negative to any hormones, 14 cells were positive for one of the pituitary hormones and/or PIT1, confirming the presence of all types of hormone-producing cells. Notably, GFP-fluorescence appeared in two hormone-positive cells during culture. On the other hand, we observed hormone-producing cells that were not positive for S100ß at the end of the time-lapse study, despite being initially positive. These findings suggest that S100ß-positive cells cultured from the anterior lobe are capable of developing into hormone-producing cells, although this happens relatively infrequently.


Subject(s)
Cell Differentiation , Green Fluorescent Proteins/metabolism , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/metabolism , Pituitary Hormones, Anterior/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Animals , Cell Count , Cell Division , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Rats, Transgenic , SOXB1 Transcription Factors/metabolism , Time-Lapse Imaging , Trypsin/metabolism
18.
J Reprod Dev ; 60(4): 304-11, 2014.
Article in English | MEDLINE | ID: mdl-24881871

ABSTRACT

Paired-related transcription factors, PRRX1 and PRRX2, which are present in mesenchymal tissues and participate in mesenchymal cell differentiation, were recently found in the stem/progenitor cells of the pituitary gland of ectodermal origin. To clarify the role of PRRX1 and PRRX2 in the pituitary gland, the present study first aimed to identify transcription factors that regulate Prrx1 and Prrx2 expression. A promoter assay for the upstream regions of both genes was performed by co-transfection of the expression vector of several transcription factors, many of which are frequently found in the pituitary stem/progenitor cells. The results for the promoter activity of both genes showed expression in a cell type-dependent manner. Comprehensive comparison of transcriptional activity of several transcription factors was performed with CHO cells, which do not show Prrx1 and Prrx2 expression, and the results revealed the presence of common and distinct factors for both genes. Among them, KLF6 showed specific and remarkable stimulation of Prrx2 expression. In vitro experiments using an electrophoretic mobility shift assay and siRNA interference revealed a potential ability for regulation of Prrx2 expression by KLF6. Finally, immunohistochemistry confirmed the presence of KLF6 in the SOX2/PRRX2 double-positive stem/progenitor cells of the postnatal pituitary gland. Thus, the finding of KLF6 might provide a novel clue to clarify the maintenance of stem/progenitor cells of the postnatal pituitary gland.


Subject(s)
Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Pituitary Gland/metabolism , Proto-Oncogene Proteins/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , CHO Cells , Cricetulus , Gene Knockdown Techniques , Immunohistochemistry , Kruppel-Like Factor 6 , Kruppel-Like Transcription Factors/genetics , Mice , NIH 3T3 Cells , Pituitary Gland/cytology , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Rats, Wistar , Transcription, Genetic
19.
J Reprod Dev ; 60(4): 295-303, 2014.
Article in English | MEDLINE | ID: mdl-24881870

ABSTRACT

The pituitary is an important endocrine tissue of the vertebrate that produces and secretes many hormones. Accumulating data suggest that several types of cells compose the pituitary, and there is growing interest in elucidating the origin of these cell types and their roles in pituitary organogenesis. Therein, the histogenous cell line is an extremely valuable experimental tool for investigating the function of derived tissue. In this study, we compared gene expression profiles by microarray analysis and real-time PCR for murine pituitary tumor-derived non-hormone-producing cell lines TtT/GF, Tpit/F1 and Tpit/E. Several genes are characteristically expressed in each cell line: Abcg2, Nestin, Prrx1, Prrx2, CD34, Eng, Cspg4 (Ng2), S100ß and nNos in TtT/GF; Cxcl12, Raldh1, Msx1 and Twist1 in Tpit/F1; and Cxadr, Sox9, Cdh1, EpCAM and Krt8 in Tpit/E. Ultimately, we came to the following conclusions: TtT/GF cells show the most differentiated state, and may have some properties of the pituitary vascular endothelial cell and/or pericyte. Tpit/F1 cells show the epithelial and mesenchymal phenotypes with stemness still in a transiting state. Tpit/E cells have a phenotype of epithelial cells and are the most immature cells in the progression of differentiation or in the initial endothelial-mesenchymal transition (EMT). Thus, these three cell lines must be useful model cell lines for investigating pituitary stem/progenitor cells as well as organogenesis.


Subject(s)
Cell Line/cytology , Pituitary Gland/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Profiling , Mice , Pituitary Gland/metabolism , Transcription Factors/metabolism
20.
J Reprod Dev ; 60(4): 288-94, 2014.
Article in English | MEDLINE | ID: mdl-24804940

ABSTRACT

Homeobox transcription factors are known to play crucial roles in the anterior lobe of the pituitary gland. During molecular cloning with the Yeast One-Hybrid System using a 5'-upstream region of the porcine Fshß as a bait sequence, we have cloned a cDNA encoding a partial sequence of the retina-derived POU domain factor 1 (RPF1) from the porcine pituitary cDNA library and confirmed its specific binding to the bait sequence. In situ hybridization was performed to examine localization of Rpf1 and showed that this gene is expressed in the stem/progenitor cells of the rat pituitary primordium as well as the diencephalon and retina. In addition, real-time PCR demonstrated that Rpf1 transcripts are abundant in early embryonic periods but that this is followed by a decrease during pituitary development, indicating that this factor plays a role in differentiating cells of the pituitary. The transcriptional activity of RPF1 for genes of Prop1, Prrx1 and Prrx2, which were characterized as genes participating in the pituitary stem/progenitor cells by our group, was then examined with full-length cDNA obtained from the rat pituitary. RPF1 showed regulatory activity for Prop1 and Prrx2, but not for Prrx1. These results indicate the involvement of this retina-derived factor in pituitary development.


Subject(s)
POU Domain Factors/metabolism , Pituitary Gland/embryology , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Gene Expression Regulation , Gene Library , In Situ Hybridization , Molecular Sequence Data , Pituitary Gland/metabolism , Rats , Rats, Wistar , Sequence Analysis, DNA , Swine
SELECTION OF CITATIONS
SEARCH DETAIL